RNA Binding Protein Syncrip Regulates the Leukemia Stem Cell Program

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 739-739
Author(s):  
Ly P. Vu ◽  
Camila Prieto ◽  
Elianna Amin ◽  
Gerard Minuesa ◽  
Sagar Chhangawala ◽  
...  

Abstract RNA binding proteins (RBPs) tightly control mRNA abundance, stability and translation while mutations or altered expression of specific factors can drive malignancy. Yet, the identity of the RBPs that govern myeloid stem cells remains poorly characterized. We and others have recently demonstrated that MUSASHI-2 (MSI2) is a central regulator of the cancer stem cell program in myeloid leukemia. Therefore, we curated a list of 127 MSI2 direct protein interactors and associated genes to perform an in vivo shRNA screen using MLL-AF9 leukemia cells. We identified shRNAs corresponding to 24 genes that were significantly depleted in vivo after sequencing and comparing their representation from day 16 to day 0. We confirmed knockdown and demonstrated marked reduction in myeloid colony formation in vitro after depleting 7 hits identified in our screen. Additionally, we tested these genes in normal bone marrow c-Kit positive cells and found that the most differentially required gene in leukemia cells compared to normal cells was SYNCRIP (Synaptotagmin-binding, cytoplasmic RNA-interacting protein). SYNCRIP is an RNA binding protein that has been implicated in various RNA regulatory processes but its role in the hematopoietic system is virtually unknown. Depletion of SYNCRIP with shRNAs in murine MLL-AF9 leukemia cells resulted in an increase in myeloid differentiation, apoptosis and delayed leukemogenesis in vivo (median survival of 35 days; control versus 61 days shRNA#1 knockdown was selected against, and "not reached" shRNA#2). To further assess SYNCRIP function in vivo, we developed a germline Syncrip knockout (KO) by injecting Cas9-DNA and Syncrip - guides RNAs into embryos and harvested E13 fetal liver cells. After Syncrip deletion was verified by immunoblotting, we observed normal numbers of HSCs and equivalent engraftment in lethally irradiated animals in both primary and secondary transplants. In contrast, we observed a delay in leukemeogenesis (median survival of 87.5 days; WT versus 118 days KO) in recipient mice after transplantation of MLL-AF9 transformed LSKs. Notably, non-deleted leukemia cells outcompeted the SYNCRIP deleted cells based on a reemergence of SYNCRIP expression. These data suggest that SYNCRIP is differentially required in myeloid leukemia cells compared to normal cells. Furthermore, we found that SYNCRIP was highly expressed in wide variety of human AML cell lines and in primary AML patients (n=4/5). SYNCRIP depletion with shRNAs resulted in reduced cell proliferation and the induction of apoptosis in human AML cell lines (MOLM13, NOMO-1, KASUMI-1 and NB4) and a marked decrease in engraftment of primary AML patient cells. To gain insights into SYNCRIP function, we performed RNA-sequencing of leukemia cells depleted for SYNCRIP. Gene set enrichment analysis (GSEA) negatively enriched for the MLL-AF9, HOXA9 and stem cell programs in SYNCRIP-KD cells and positively enriched for MSI2's direct mRNA binding targets and a MSI2 deficient LSC signature. Reciprocal immunoblotting in the presence or absence of RNAse demonstrated that SYNCRIP and MSI2 interaction is RNA dependent. We validated their shared targets by performing SYNCRIP RNA-immunoprecipitation (RIP) for previously identified MSI2's direct mRNAs targets (HOXA9 and c-MYC). SYNCRIP depletion resulted in reduced protein abundance of HOXA9 and c-MYC. Forced MSI2 expression partially rescued the colony formation and HOXA9 expression in SYNCRIP-KD cells. To assess the functional downstream targets of SYNCRIP in leukemia, we overexpressed HOXA9 and c-MYC in SYNCRIP-KD cells and observed that HOXA9 expression but not c-MYC partially rescues the effect of SYNCRIP depletion on myeloid colony formation. Mechanistically, we showed that SYNCRIP regulates translation of HOXA9 without affecting HoxA9 mRNA stability. Overall, we provide a strategy for interrogating the functional RNA binding network in leukemia using shRNA screening. Additionally, we validated SYNCRIP as a novel RBP that controls the leukemia stem cell program and propose that targeting these functional complexes might provide a novel therapeutic strategy in myeloid leukemia. Disclosures Melnick: Janssen: Research Funding. Levine:Novartis: Consultancy; Qiagen: Membership on an entity's Board of Directors or advisory committees. Järås:Cantargia AB: Equity Ownership.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 883-883
Author(s):  
Camila Prieto ◽  
Diu Nguyen ◽  
Ly P Vu ◽  
Alexendar Perez ◽  
Saroj Gourkanti ◽  
...  

Abstract Acute myeloid leukemia (AML) is characterized by a block in the development of myeloid cells, often due to dysregulation of genes involved in key processes including self-renewal, proliferation, and differentiation. Somatic mutations and aberrant expression of RNA binding proteins (RBPs) have recently been found to be important in hematological malignancies. For example, our group and others have recently determined that increased expression of MUSASHI-2 and SYNCRIP drives aggressive leukemia. To discover novel RBP regulators of leukemia, we performed an in vivo pooled shRNA screen of 127 MSI2 direct protein interactors and associated genes (Vu et al. Nat Gen. 2017). In this screen, shRNAs specific to the RBP RBMX (RNA binding motif protein, X-linked) were selectively depleted in murine MLL-AF9 driven leukemia. RBMX has been implicated in regulating alternative splicing, chromatin cohesion, and DNA-damage response, but its function in hematopoiesis and leukemia is not known. We confirmed that depletion of RBMX with shRNAs in murine MLL-AF9 leukemia cells resulted in reduced myeloid colony formation, increased apoptosis, and increased differentiation as determined by flow analysis of myeloid cell surface markers Gr-1 and Mac-1 (mean of 61-65% shRNA versus mean of 12.95% control). Furthermore, RBMX is highly expressed among human myeloid leukemia cell lines (n=10/11) and primary AML patient samples (n=2/4). Depletion of RBMX with shRNAs led to a dramatic decrease in cell proliferation and 3-fold induction of apoptosis in several human myeloid leukemia cell lines (MOLM-13, THP-1, K562, and KCL-22). Additionally, RBMX depletion in AML cells induced myeloid differentiation and significantly delayed leukemogenesis cells in vivo (median survival of 51.5 days in control versus median 'not reached' in shRNA1 and shRNA2). To determine if there is a differential requirement of RBMX in survival of leukemia cells compared to normal hematopoietic stem and progenitor cells (HSPCs), we depleted RBMX with shRNAs in normal murine bone marrow c-Kit+ cells and found no significant changes in colony formation. Depleting RBMX with shRNAs in human cord blood derived CD34+ HSPCs resulted in reduced colony formation but no increase in apoptosis. Thus, these data suggest that there is a differential requirement for RBMX in myeloid leukemia cells compared to normal cells. To uncover the mechanism of RBMX function, we performed RNA-sequencing of human AML cells (MOLM-13) depleted for RBMX. Gene set enrichment analysis demonstrated a loss of cell cycle and DNA repair associated programs in RBMX depleted cells. Complex chromosomal karyotyping analysis of these cells revealed increased metaphases with breaks and gaps (mean of 30.67% shRNA versus mean of 13.33% control) and irregular chromatin compaction (mean of 47.67%shRNA versus mean of 20% control), while cell cycle analysis showed significantly increased S-phase arrest as determined by flow analysis of Hoechst stained cells (mean of 37-40% shRNA versus of 24.18% control). Reanalysis of RBMX transcriptome-wide binding sites in 293T cells revealed that RBMX directly binds to heterochromatin protein HP1α transcripts (Liu et al. Nucleic Acids Res. 2017). HP1α, also called CBX5, is a key heterochromatin protein that binds to histone H3-K9 tri-methylation marks to promote heterochromatin formation, which is critical in chromatin condensation and chromosome segregation. HP1α has also been determined to be required for MLL leukemia stem cell maintenance. We demonstrated that RBMX depletion resulted in a significant decrease of HP1α mRNA expression without affecting its mRNA stability in AML cells. We confirmed that RBMX depletion reduced the protein abundance of HP1α. Moreover, overexpression of HP1α rescued the effect of RBMX depletion on cell growth and apoptosis. Our study finds that RBMX binds to HP1α mRNA and regulates the transcriptional activity of the HP1α locus, which then maintains proper chromatin compaction in leukemia cells. Overall, we determine that RBMX function is critical for myeloid leukemia survival and has potential as a novel therapeutic target in AML. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 3-3
Author(s):  
Xiaomin Wang ◽  
Tengxiao Guo ◽  
Yuxia Wang ◽  
Shengnan Yuan ◽  
Shuaibing Hou ◽  
...  

Acute myeloid leukemia (AML) is an aggressive form of hematologic malignancies, caused by the accumulation of immature leukemic blasts in the hematopoietic system. Current chemotherapy regimens remain ineffective as assessed by low remission rate and 5-year-survival rate. MLL-AF9 mutations occur in approximately 10% of all acute leukemia patients, and are associated with poor therapy response and prognosis. Deregulated expression of ELAV-like family protein 2 (CELF2) has been identified in AML patients with MLL-AF9. As an RNA binding protein, CELF2 has been shown to regulate RNA splicing and embryonic hematopoietic development. However, the role of CELF2 in AML progression is still largely unknown. In this study, we first analyzed genetic data of 160 AML cases in the Cancer Genome Atlas in which CELF2 deletion account for 10% of the total cases. The survival time of patients with CELF2 deletion was much shorter than that of the others (P < 0.001). Interestingly, the expression of CELF2 was significantly decreased in different sub-type of acute myeloid leukemia cells, and the lowest expression of CELF2 was seen in MLL-AF9 (MA9) AML cells, when compared with that of normal myeloid cells. These results suggested that CELF2 deletion/mutation may play an important regulatory role in MA9-induced AML progression. To evaluate the potential role of Celf2 in the hematopoietic malignant transformation, we generated hematopoietic specific Celf2-deficient mice (Vav1-Cre;Celf2fl/fl) and found that Celf2-deficient hematopoietic stem cells (HSCs) over-expanded upon transplantation indicating a role of Celf2 in HSC proliferation. Consequently, the percentage of Celf2-KO HSC-derived myeloid cells was significantly increased in peripheral blood and bone marrow of recipient mice when compared with that of control mice. Celf2-KO Lin- cells were then infected by MA9-containing retrovirus and transplanted into lethally irradiated mice to generate Celf2-KO+MA9 mice. All Celf2-KO+MA9 mice succumbed to AML in 35 days, and their survival time is shorter than that of Celf2-WT+MA9 mice. The Celf2-KO+MA9 mice showed more aggressive phenotypes of AML than Celf2-WT+MA9 mice, including higher counts of WBCs, neutrophils and lymphocytes in peripheral blood, as well as higher extramedullar infiltration in spleen, liver and lung. To quantify the functional LSCs in Celf2-KO+MA9 mice and control MA9 mice, extreme limiting dilution transplantation assay was performed. The penetrance rate of leukemia in mice that received 1x103Celf2-KO+MA9 cells was significantly higher than that of control mice (100% versus 50%). The frequency of functional LSCs was significantly increased in Celf2-KO+MA9 mice as compared to that of the control, supporting a role of loss of Celf2 in promoting leukemia blast self-renewal. To investigate the underlying molecular mechanisms of Celf2 deletion in accelerating MA9 AML progression, we performed RNAseq to analyze the transcriptome programing changes associated with Celf2 in isogenic Celf2-WT+MA9 or Celf2-KO+MA9 AML cells. Consistent with severe extramedullar infiltration of leukemia cells in Celf2-KO+MA9 mice, gene set enrichment analysis showed up-regulated leukocyte activation and migration in Celf2-KO+MA9 leukemia cells. We next analyzed the downstream signaling pathways targeted by Celf2, and found that the phosphorylation of S6 was increased in Celf2-KO+MA9 cells, suggesting that Celf2 deletion might promote leukemia progression by activating mTORC1 signaling pathway. To confirm this, we treated Celf2-KO+MA9 mice with MA9 inhibitor (EPZ5676) and mTORC1 inhibitor (Rapamycin), and found a collaborative response to the treatment with a significant attenuation in leukemia burden in vivo. In conclusion, we found that CELF2 low expression is associated with AML patients with MA9 mutation. Celf2 deletion in mice induced myeloid-biased differentiation of HSCs. Notably, Celf2 deficiency promoted MA9-induced AML progression by activating mTORC1 signaling pathway independent of MA9 signaling pathway. Combinational therapy with MA9 inhibitors and mTORC1 inhibitors could significantly decrease the Celf2-KO+MA9 leukemia progression in vivo, indicating that combinational usage the drugs may have synergistic benefit for MA9 AML patients with low CELF2 expression. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 15-15
Author(s):  
Diu T.T. Nguyen ◽  
Camila Prieto ◽  
Zhaoqi Liu ◽  
Justin Wheat ◽  
Alexendar Perez ◽  
...  

Mutations and aberrant expression of RNA binding proteins (RBPs) have recently been found to contribute to leukemia development (Prieto and Kharas, CSH, 2020). Previously we have identified the RBP RBMX (RNA binding motif protein, X-linked) in our shRNA in vivo screen using murine MLL-AF9 driven leukemia (Prieto et al, ASH abstract 2018). Here we uncover the role of RBMX and its retrogene RBMXL1 in normal hematopoiesis and leukemogenesis in mouse and human systems. To investigate RBMX function in normal hematopoiesis, we created an Rbmx Mx1-Cre conditional knockout model to specifically delete Rbmx in the hematopoietic system. We gender stratified our studies since Rbmx is sex-linked. Deletion of Rbmx in both female and male primary mice as well as in non-competitive transplant animals did not affect hematopoiesis. Additionally, Rbmx knockout (KO) leads to a mild reduction in multipotent progenitors (MPP2 and MPP4) in female competitive transplanted mice although no defects in long-term hematopoiesis was observed in male competitive transplanted mice. These data suggest that Rbmx is dispensable for normal hematopoiesis. To identify the role of RBMX in leukemogenesis we knockout Rbmx in MLL-AF9 murine leukemia cells and found it significantly reduced colony formation in vitro and delayed leukemogenesis in vivo, indicating that Rbmx is required for leukemia maintenance. We observed, however, that MLL-AF9 transformed cells from Rbmx KO donor mice showed no delay in leukemia initiation versus cells from wildtype. We then determined that while Rbmx deletion is effective with complete depletion of mRNA, the retrogene RBMXL1 expression is maintained in Rbmx deficient cells, which may compensate for Rbmx deletion in leukemia initiation. Indeed, depletion of RBMXL1 by shRNAs (KD) in Rbmx deficient leukemia cells results in a drastic reduction in colony formation, increased colony myeloid differentiation, and induced apoptosis in cells deficient for both RBMXL1 and RBMX compared to those only depleted of RBMX. Correspondingly, RBMXL1 KD in Rbmx deficient leukemia cells resulted in further delayed leukemogenesis in vivo, indicating that RBMXL1 is functionally redundant to RBMX and both genes are required for leukemia development and maintenance. We next investigated the role of RBMX/L1 in normal human hematopoietic and leukemia cells. We found that RBMX/L1 expression were higher in AML cell lines (n=10/11) and primary AML patient samples (n=2/4) compared to healthy individuals. RBMX/L1 KD by shRNAs led to a dramatic decrease in cell proliferation, induction of apoptosis and myeloid differentiation in several human myeloid leukemia cell lines (MOLM-13, THP-1, K562, and KCL-22). Additionally, RBMX/L1 depletion significantly delayed leukemogenesis in vivo of AML cell lines (median survival of 51.5 days in control vs. median 'not reached' in shRNA1 and shRNA2), and of primary leukemia cells derived from an AML patient (median 50 days in control vs. "not reached" in shRNA2). We next depleted RBMX/L1 in human CD34+ stem and progenitor cells and observed reduced colony formation but no increase in apoptosis. Taken together, our data suggest that RBMX/L1 are differentially required in leukemia cells versus normal cells in both human and mouse systems. To uncover the mechanism of RBMX/L1 function, we performed complex chromosomal karyotyping analysis of RBMX/L1 depleted MOLM13 cells and revealed increased metaphases with breaks and gaps. In addition, ATAC-seq analysis showed profound genome-wide changes in chromatin accessibility and compaction upon RBMX/L1 depletion. Analyses of RNA-seq and transcriptome-wide RBMX/L1 binding targets (PAR-CLIP; Liu et al. 2017) revealed that RBMX/L1 directly binds to mRNAs and affects transcription of multiple loci including the heterochromatin protein 1 alpha (HP1a) mRNA. Using single molecule RNA FISH, we uncovered that RBMX/L1 controls the nascent transcription of the HP1a locus. Forced HP1a expression rescued the RBMX/L1 depletion effects on cell growth and apoptosis. Overall, we determine that RBMX/L1 control leukemia cell survival by regulating chromatin state through its downstream target HP1a. This study describes a novel genetic compensation phenomenon in leukemia and illuminate a mechanism for RBPs directly promoting transcription. Our results suggest RBMX/L1 as well as HP1a as potential novel therapeutic targets in myeloid malignancies. Disclosures Steidl: Aileron Therapeutics: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Stelexis Therapeutics: Consultancy, Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Bayer Healthcare: Research Funding; Pieris Pharmaceuticals: Consultancy. Kharas:Accent Therapeutics: Consultancy; 28-7: Research Funding.


Blood ◽  
1992 ◽  
Vol 80 (5) ◽  
pp. 1199-1206 ◽  
Author(s):  
T Pietsch ◽  
U Kyas ◽  
U Steffens ◽  
E Yakisan ◽  
MR Hadam ◽  
...  

Abstract A novel hematopoietic growth factor, the stem cell factor (SCF), for primitive hematopoietic progenitor cells has recently been purified and its gene has been cloned. In this study we tested the mitogenic activity of recombinant human SCF on myeloid leukemia cells as well as the expression of its receptor. We have investigated the proliferation of 31 myeloid leukemia cell lines as well as fresh myeloid leukemic blasts from 17 patients in a 72-hour 3H-thymidine uptake assay in the presence of various concentrations of recombinant human (rh) SCF alone or in combination with saturating concentrations of granulocyte- macrophage colony-stimulating factor (GM-CSF), G-CSF, M-CSF, interleukin-3 (IL-3), or erythropoietin (EPO). Only five of 31 lines, but fresh leukemic blasts from 12 of 17 patients with acute myeloid leukemia (AML), significantly responded to SCF. The responding cell lines were of the acute promyelocytic, chronic myeloid, megakaryoblastic, and erythroleukemia origin, the responding blast preparations of all French-American-British subtypes. Synergistic activities of SCF were found with G-CSF, GM-CSF, EPO, and IL-3. To determine the SCF binding sites on leukemic cells, we used 125I- radiolabeled SCF in Scatchard analysis and cross-linking studies. The leukemic cell lines responding to SCF expressed from 2,300 up to 29,000 binding sites per cell. The SCF receptor expression was downregulated in vitro by the presence of its ligand. Cross-linking studies demonstrated a 150-Kd SCF receptor on the surface of all responding myeloid leukemias. This study suggests that SCF may be an important factor for the growth of myeloid leukemia cells, either as a direct stimulus or as a synergistic factor for other cytokines. Furthermore, using polymerase chain reaction analysis of total RNA from the myeloid leukemia lines, we found expression of SCF-mRNA in 17 of 30 lines, suggesting autocrine mechanisms in the growth of a subgroup of leukemic cells by coexpression of SCF and its receptor.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2531-2531
Author(s):  
Li-Yuan Bai ◽  
Jing-Ru Weng ◽  
Chia-Yung Wu ◽  
Chang-Fang Chiu ◽  
Su-Peng Yeh ◽  
...  

Abstract Introduction Indole-3-carbinol (I3C) is a broadly targeted phytochemical shown to prevent carcinogenesis in animal studies and to suppress the proliferation of cancer cells of human breast, colon, prostate, and endometrium. Here we aim to test the anticancer effect of OSU-A9, an I3C derivative with improved potency, in acute myeloid leukemia (AML). Materials and Methods The in vitro activity of OSU-A9 was evaluated in AML cell lines (HL-60 and THP-1) and primary leukemia cells from 18 AML patients. THP-1 xenograft tumors in athymic nude mice was used for in vivo study. Results OSU-A9 mediates cytotoxicity in AML cell lines and primary leukemia cells from AML patients in a dose-responsive manner. The IC50 at 24 h for 18 patients was 1.63 μM. Normal human bone marrow cells were much less sensitive to OSU-A9 with an IC50 at 24 h greater than 8 μM. OSU-A9 causes cytotoxicity dependent on caspase activation, as evidenced by caspase-3 and PARP cleavage, and induces autophagy but not autophagic cell death. Interestingly, pretreatment of AML cell lines and primary AML cells with N-acetylcysteine or glutathione rescues them from apoptosis (and concomitant PARP cleavage) and Akt hypophosphorylation, implicating a key role of reactive oxygen species (ROS) in OSU-A9-related cytotoxicity. To investigate the anti-leukemia effect of OSU-A9 in vivo, fifteen male athymic nude mice were xenografted with THP-1 cells. Briefly, the anticancer utility of OSU-A9 is extended in vivo as it, administered intraperitoneally, suppresses the growth of THP-1 xenograft tumors in athymic nude mice without obvious toxicity. For biomarker analysis in the THP-1 xenografts, protein extracts were obtained from the tumors and immunoblotted for Akt levels. The tumors from OSU-A9 treated mice exhibited down regulation of Akt phosphorylation compared with those from placebo-controlled mice. Conclusions This study shows that ROS-mediated apoptosis contributes to the anticancer activity of OSU-A9 in AML cell lines and primary AML cells, and thus should be considered in the future assessment of its translational value in AML therapy. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 133 (18) ◽  
pp. 1935-1953 ◽  
Author(s):  
Shuming Wang ◽  
Yilin Hu ◽  
Xiurui Lv ◽  
Bin Li ◽  
Dianhua Gu ◽  
...  

Abstract Circular RNAs (circRNAs) play a vital role in cancers. Accumulated evidences showed that the physiological condition of cells can be reflected by the circRNAs in the exosomes they secrete, and these exosomal circRNAs can be captured by the receptor cells, thereby inducing a series of cellular responses. We performed qRT-PCR to detect the expression level of circ-0000284 in cholangiocarcinoma cell lines, tissues and plasma exosomes. Then the direct interaction between circ-0000284 and miR-637 was investigated through dual-luciferase reporter assay, RNA binding protein immunoprecipitation (RIP) assay and Fluorescent in situ hybridization (FISH) assay. Subsequently, EdU (5-ethynyl-2′-deoxyuridine), migration, invasion assay, flow cytometry and nude mouse tumorigenicity assay were adopted to evaluate the effect of circ-0000284 on migration, invasion, proliferation and apoptosis of cholangiocarcinoma cells. Additionally, TEM was conducted to investigate the shape and size of exosomes from cholangiocarcioma and 293T cell lines. Circ-0000284 was evidently elevated in cholangiocarcinoma cell lines, tumor tissues and plasma exosomes. Meanwhile, the high expression of circ-0000284 enhanced the migration, invasion and proliferation abilities of cholangiocarcinoma cells in vivo and in vitro. Besides, the levels of circ-0000284 were increased in cholangiocarcinoma cells and exosomes from them. Moreover, exosomes from cholangiocarcinoma cells enhanced circ-0000284 expression and stimulated migration and proliferation of the surrounding normal cells. Our findings suggest that on the one hand circ-0000284 functions as a competitive endogenous RNA to promote cholangiocarcinoma progression, and on the other hand, circ-0000284 can be directly transferred from cholangiocarcinoma cells to surrounding normal cells via exosomes and in this way regulate the biological functions of surrounding normal cells.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1010-1010
Author(s):  
Noriko Sugino ◽  
Masahiro Kawahara ◽  
Takayoshi Suzuki ◽  
Yuya Nagai ◽  
Yayoi Shimazu ◽  
...  

Abstract Background: Histone methylation is one of the major systems of epigenetics and reversibly regulated by lysine (K) specific methyltransferases (KMTs) and demethylases (KDMs). Dysregulation of KMTs such as EZH2 and MLL play a key role in the pathogenesis of acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) while the precise function of KDMs remains unclear.KDM1A is the first reported histone demethylase, which mainly catalyzes demethylation of mono- and di-methylated lysine 4 of histone 3 (H3K4me1 and me2 respectively). According to recent reports, the inhibition of KDM1A either alone or in combination of all-trans retinoic acid is effective for AML expressing MLL-AF9 or for several types of AML respectively, suggesting KDM1A could be a therapeutic target of AML. However, other reports argue that KDM1A is essential in hematopoiesis, raising concern that KDM1A-targeted therapy could lead to severe hematological toxicity. Here, we try to clarify what types of leukemia can be ameliorated by the pharmacological inhibition of KDM1A with a possible therapeutic window and what are functional and molecular mechanisms utilizing highly selective KDM1A inhibitors we have newly designed. Results: First we validated the effect of our novel inhibitors on murine leukemia cells harboring MLL-AF9. In accordance with a previous report, our novel KDM1A inhibitors suppressed cell proliferation, diminished clonogenic capacity and induced G1-S cell cycle arrest and myelomonocytic differentiation but not apoptosis in quite low concentration that clonogenicity of normal murine bone marrow cells was spared. Next we examined the effect of these drugs on diverse types of human myeloid leukemia cells and found that our drugs were particularly effective in erythroid leukemia cells (HEL), megakaryocytic leukemia cells (CMK11-5), and a blastic subline from a MDS patient with complex karyotype (MDS-L). MDS-L cells were changed phenotypically and morphologically towards myelomonocytic differentiation such as the increase of CD11b expression level and the induction of neutrophil-like cells. HEL and CMK cells which are negative for myelomonocytic markers also gained CD11b expression and decreased erythroid markers such as CD235a and CD71. These data suggest that the inhibition of KDM1A induces myeloid differentiation across various types of leukemia. To investigate an underlying molecular basis for the cell fate conversion in HEL and myeloid differentiation in MDS-L by the inhibition of KDM1A, we performed gene expression profiling and analyzed a change of gene signatures. The expression pattern of transcriptional factors was changed from the erythroid signature (e.g. GATA1 and TAL1) to the myeloid signature (e.g. SPI1 and CEBPA) in HEL. Gene Set Enrichment Analysis (GSEA) showed that the myeloid differentiation-associated gene signature was positively enriched and the leukemia stem cell-associated gene signature was negatively enriched in both HEL and MDS-L. Finally, we investigated the effect of our KDM1A inhibitors on primary human samples such as AML with MLL-AF9 and MDS in the phase of overt leukemia (MDS/AML) with complex karyotype. The colony formation capacity was clearly impaired in relatively low concentration that normal colonies were spared. We transplanted primary MDS/AML cells with complex karyotype to immunodeficient mice and treated with a KDM1A inhibitor or vehicles after confirming the engraftment. In one MDS/AML case, all mice treated with vehicles (n=4) died of anemia and the increase of human leukemia cells within four months while 3 of 4 mice treated with a KDM1A inhibitor have survived for more than six months. Also in another case, both of two vehicle-treated mice died while all drug-treated mice had survived for more than six months. At day 200 after transplantation, we sacrificed all survived mice treated with a KDM1A inhibitor and found that human blasts were displaced from the bone marrow of treated mice. Those data suggest that our KDM1A inhibitor is effective in vivo and have a possibility for the clinical application. Conclusion: Our study suggests that KDM1A involves with myeloid differentiation and the leukemia stem cell signature and that the pharmacological inhibition of KDM1A by highly selective inhibitors is a promising way to ameliorate AML with poor prognosis such as erythroleukemia and MDS/AML with complex karyotype, without impairing normal hematopoiesis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. SCI-3-SCI-3
Author(s):  
Tannishtha Reya

Our research focuses on the signals that control stem cell self-renewal and how these signals are hijacked in cancer. Using a series of genetic models, we have studied how classic developmental signaling pathways play key roles in hematopoietic stem cell growth and regeneration and are dysregulated during leukemia development. Through this work we have identified Hedgehog and Wnt signaling, and more recently the cell fate determinant Musashi, as critical players in driving progression of hematologic malignancies and as targets for therapy. To search for new regulators of myeloid leukemia, we have carried out a focused screen of surface molecules that may enable leukemia cells to receive supportive cues from the microenvironment. This screen identified key new adhesion signals that are critical to leukemia growth, drug resistance and dissemination. Using high resolution in vivo imaging we have mapped how these mediate the interactions that leukemia cells make within their microenvironment. To complement this focused screen, we have also carried out a genome wide CRISPR screen to more generally define the biological determinants of myeloid leukemia establishment and propagation. This screen identified a large number of new genes and programs critically required for leukemia, including those essential for chromatin remodeling and spliceosomal assembly. Among these, RNA binding proteins (RBPs) in general, and the chromatin binding sub-family of RBPs in particular, emerged as key new dependencies of myeloid leukemia. The talk will focus in part on these new regulators. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3936-3936
Author(s):  
Yongwei Su ◽  
Xinyu Li ◽  
Holly Edwards ◽  
Lisa Polin ◽  
Juiwanna Kushner ◽  
...  

Abstract Although standard induction therapy initially elicits a promising response in the majority of acute myeloid leukemia (AML) patients, the majority relapse. Leukemia stem cells (LSCs) that survive chemotherapy are believed to be responsible for AML relapse. Therefore, new therapies that eliminate LSCs are desperately needed. ONC201 is a TRAIL inducer and the founding member of the imipridone family. It has been shown to induce apoptosis in LSCs (Ishizawa et al, Science Signaling. 2016; 9:ra17). ONC201 was chemically modified to increase the potency and selectivity against cancer cells, resulting in the new analog ONC213. In this study, we investigated the antileukemic activity and the underlying molecular mechanism of ONC213 in preclinical AML models. ONC213 activity in AML cell lines and primary AML patient samples was first tested in vitro. MTT assay results revealed that ONC213 IC50s ranged from 91.7 nM to 2.4 µM in AML cell lines and primary AML patient samples, which are achievable in vivo based on results from a PK study in mice (a single dose of 50 and 100 mg/kg ONC213 resulted in peak plasma concentrations of 3.7 μM and 8 μM, respectively). Annexin V/propidium iodide staining and flow cytometry analysis results showed variable responses for the AML cell lines tested. After 48 h treatment with 500 nM ONC213, striking induction of cell death in MOLM-13 and MV4-11 cells was detected (at least 72% Annexin V+ cells), while THP-1 and U937 cells showed little to no increase in Annexin V+ cells (6-11%). Similar results were obtained in primary AML patient samples. In contrast to the 48 h treatment of THP-1 and U937 cells, increasing the treatment duration to 120 h resulted in greater than 50% Annexin V+ cells, suggesting that a longer exposure time is necessary in some cell lines. In MV4-11 and MOLM-13 cells, initiation of cell death was detected 8 to 12 h post ONC213 treatment. Colony formation assays revealed that ONC213 treatment significantly reduced colony formation capacity of primary AML patient samples to less than 5% compared to vehicle control, while having no significant effect on normal hematopoietic progenitor cells. A primary AML patient sample was treated with or without ONC213 for 48 h, transplanted into NSG mice, and ten weeks later bone marrow was harvested and human CD45+ cells were measured. ONC213 treatment significantly reduced human AML engraftment compared to vehicle control (0.6% vs. 21.3%; p<0.05), demonstrating that ONC213 kills LSCs in vitro. Next, we examined in vivo efficacy of ONC213 against an AML cell line derived xenograft mouse model. MV4-11 cells were injected into NSGS mice through the tail vein. Three days post-injection, the mice were randomized into vehicle control or 125 mg/kg ONC213 cohorts (5 mice per cohort) and treated daily for 8 days. Modest weight loss was noted but was entirely manageable. ONC213 treatment extended the survival of mice by 88% (median survival 62 vs 33 days). Unlike ONC201, ONC213 treatment of AML cells did not increase the expression of TRAIL. Interestingly, RNAseq results showed that 500 nM ONC213 treatment for 48 h downregulated 33 mRNAs in the oxidative phosphorylation (OXPHOS) pathway, suggesting that ONC213 treatment decreases OXPHOS in AML cells. Thus far, six of the downregulated mRNAs (UQCRQ, SDHA, COX6C, NDUFS5, ATP5D, and NDUFB1) were verified by real-time RT-PCR after both 8 h and 48 h ONC213 treatment. LSCs have been shown to be highly reliant on OXPHOS, while normal hematopoietic stem cells and some bulk AML cells can switch to glycolysis for ATP production during times of OXPHOS inhibition. Thus, ONC213 may kill LSCs through inhibition of OXPHOS. In addition to downregulation of OXPHOS related genes, we found that ONC213 treatment downregulates Mcl-1. Since Mcl-1 mediates resistance to the promising Bcl-2-selective inhibitor ABT-199 (Venetoclax) and inhibition of Bcl-2 impairs OXPHOS, ONC213 would likely synergize with ABT-199 in AML cells. Indeed, combined treatment resulted in striking synergistic induction of apoptosis in both AML cell lines and primary patient samples. Enhanced cell death was detected 8 h post combination treatment in both MOLM-13 and MV4-11 cells. Results from colony formation assays revealed that the combination spares normal hematopoietic progenitor cells. Taken together, ONC213 is active as a single agent and in combination with ABT-199 in AML. Disclosures Allen: Oncoceutics: Employment. Stogniew:Oncoceutics: Employment. Prabhu:Oncoceutics: Employment. Ge:MEI Pharma: Research Funding.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 791-791
Author(s):  
Ly P Vu ◽  
Brian F Pickering ◽  
Yuanming Cheng ◽  
Zaccara Sara ◽  
Diu Nguyen ◽  
...  

Abstract N 6-methyladenosine (m6A) is a nucleotide modification in mRNA that is required for the acquisition of cell fate in embryonic stem cells. Recent studies have indicated that methylation writers can act as both oncogenes and tumor suppressor genes. Here we show that m6A is a critical regulator of myeloid differentiation of human hematopoietic stem and progenitors and myeloid leukemia cells. Depletion of the m6A-forming enzyme METTL3 with shRNAs in human cord blood derived CD34+ hematopoietic stem/progenitor cells (HSPCs) decreased global m6A levels, promoted differentiation, and reduced cell proliferation and colony formation. Conversely, overexpression of wild type METTL3-WT, but not the catalytic dead form METTL3-CD, in HSPCs increased global m6A levels, inhibited differentiation, and promoted cell growth. We surveyed METTL3 expression across different cancers and found that METTL3 is expressed at the highest level in acute myeloid leukemia (AML) compared to a broad range of solid tumors. METTL3 was highly expressed in wide variety of human AML cell lines (11/11) and in primary AML patients (3/3). Utilizing 2D-Thin layer chromatography (TLC), we found that m6A levels in poly(A) mRNAs were significantly increased in the MOLM13 leukemia cells and primary AML patient cells compared to normal HSPCs. To directly address the role of m6A in human myeloid leukemia cells, we demonstrated that METTL3 depletion with shRNAs resulted in reduced cell proliferation and the induction of apoptosis in human AML cell lines (MOLM13, NOMO-1 and KASUMI-1), and delayed leukemia in vivo (MOLM13 cells - median survival of 23 days control versus 35 and 36 days shRNA#9 and shRNA#12 respectively - in vivo knockdown was negatively selected as determined by immunoblotting). Validating an on-target effect, expression of shRNA-resistant form of METTL3 rescued both the apoptotic and differentiation phenotypes in METTL3-depleted cells and CRISPR/Cas9 mediate depletion of METTL3 (using two independent guide RNAs) resulted in similar phenotypes. Additionally, we looked into the rank of m6A "writers" and "erasers" in the genome-wide CRISPR-based screen (Wang et al. Cell 2017) for genes essential for survival in 14 AML cell lines. Interestingly, while all members of the "writer" complexes, METTL3, METTL14, WTAP and KIAA1429, were scored highly (at the top 10%), the eraser ALKBH5 showed no essentiality and FTO is only important for survival of EOL-1 cells. This data suggests that the m6A writer complex is required for leukemia. To gain insights into the mechanism of why m6A and METTL3 are required in leukemia, we performed single-nucleotide resolution mapping of m6A in MOLM13 cells using miCLIP, gene expression analysis, ribosome profiling and reverse phase protein arrays in MOLM13 cells depleted for METTL3. We profiled mRNAs targeted for m6A modifications and found that transcripts with m6A were more stable but were translated with less efficiency. Gene Set Enrichment Analysis revealed negative enrichment of MYC and ESCs gene sets. We further validated c-MYC, BCL2 and PTEN as targets for m6A modifications. We performed meRIP-qPCR and showed that knockdown of METTL3 specifically reduced enrichment of m6A at mapped sites of these transcripts. We demonstrated that c-MYC, BCL2 and PTEN protein expression were reduced despite a 2-5 log2 fold increase in mRNA expression after METTL3 depletion. The effects were reversed in cells overexpressing METTL3-WT, but not the METTL3-CD. Importantly, we observed a robust activation of p-AKT upon METTL3 knockdown, and treatment with inhibitors of PI3K and AKT partially rescued the differentiation effects in METTL3 depleted cells. Overall, m6A is critical for maintaining the differentiation program in the hematopoietic system and that this process is dysregulated in myeloid leukemia. Our data provides a rationale for targeting the mRNA methylation program in myeloid leukemia. Disclosures Carroll: Astellas Pharmaceuticals: Research Funding; Incyte Pharmaceuticals: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document