scholarly journals Targeting PRMT9 Suppresses Acute Myeloid Leukemia Maintenance

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 358-358
Author(s):  
Haojie Dong ◽  
Xin He ◽  
Lei Zhang ◽  
Wei Chen ◽  
Yuhui Wu ◽  
...  

Abstract AML is a heterogenous disease in which prognosis and treatment is determined by recurrent genetic mutations and chromosomal abnormalities. Some genetic alterations result in aberrant RNA translation, which is exploited by leukemia stem cells (LSCs) to produce short-lived oncoproteins (c-Myc, Mcl-1) to promote cell survival. Since LSCs are considered as the source of treatment failure and relapse, it is critical to understand how LSCs hijack the translational machinery in order to develop effective therapeutics in AML. Arginine methylation catalyzed by protein arginine methyltransferases 1-9 (PRMT1-9) regulates various activities, including RNA translation. We revealed that PRMT1 over activation contributes to leukemia maintenance (Blood, 2019; Blood, 2019). Unlike PRMT1, the recently described PRMT9, is not defined in leukemia. Follow up of AML patients from existing datasets (GSE12417, TARGET) confirms that patients with higher levels of PRMT9 correlates with decreased overall survival. In our AML cohort, we observed significantly increased PRMT9 mRNA levels in AML CD34 + cells relative to normal counterparts (Fig. 1A), spurring our interest in evaluating its function in AML. We thus developed a Prmt9 conditional KO mouse (Mx1-Cre/Prmt9 f/f) and crossed it with an MLL-AF9 (MA9) knock-in mouse to generate Prmt9-KO/MA9 mice for further transplant assay. As a result, Prmt9-KO significantly delayed leukemogenesis in recipient mice carrying MA9 transplants evidenced by prolonged survival (Fig. 1B). Prmt9-KO impaired leukemia-initiating activity evidenced by secondary transplantation that significant less residual CD45.2 + MA9 leukemia cells were found in BM of secondary recipients receiving Prmt9-KO cells relative to those of Prmt9-WT controls. We next validated PRMT9 function in human AML. PRMT9-KD potentially impaired survival of primary AML CD34 + cells, whereas large sparing normal counterparts (Fig. 1C). Moreover, we designed a WT PRMT9 construct and corresponding catalytically dead mutant, both resistant to shPRMT9 (WT-R and MUT-R). Unlike MUT-R, WT-R expression rescued survival effects seen following shPRMT9 treatment, indicating that PRMT9 catalytic activity is required for AML survival (Fig. 1D). To define PRMT9 downstream effectors, we searched for methylated substrates in Molm13 with inducible PRMT9-KD or corresponding controls by performing SILAC analysis coupled with quantitative Mass-Spec (MS) (Fig. 1E). Complete analyses of normalized methyl peptides SILAC ratios revealed more prominent down-regulation of R-methylation in PRMT9-KD cells, with 49 methyl sites downregulated. Notably, methylated PAPB1 C-terminus peptide containing R493 was most significantly depleted upon PRMT9-KD. PABP1 potentiates translation initiation by binding to the poly(A) mRNA tail and interacting with factors like eIF4G. We thus generated an anti-R493 methylation antibody. Indeed, PRMT9 was confirmed to catalyze R493 methylation through in-vitro and cellular methylation assays (Fig. 1F, G). To define the function of R493 methylation, we ectopically expressed either WT PABP1 or the R493K mutant in Molm13 cells and then engineered those cells to express shPABP1 to KD endogenous PABP1. Notably, unlike WT PABP1, expression of R493K suppressed protein synthesis and increased apoptosis after endogenous PABP1-KD, suggesting that PRMT9 mediated PABP1-R493 methylation promotes AML viability (Fig. 1H, I). To discover PRMT9 inhibitors, we conducted a structure-based virtual screen of 960,000 compounds from NCI and Zinc compound libraries (Fig. 1J). We requested the top 300 candidates to assess their anti-leukemia activity in Molm13 cells (Fig. 1K). The top 20 most effective compounds were further analyzed via PRMT9 methylation assay (R493 antibody detection). NSC641396 exhibited the most potent PRMT9 inhibitory effects, as evidenced by decreased PABP1 R493 methylation levels at a dose <1 µΜ (Fig. 1L). NSC641396 treatment on AML CD34 + cells altered polysome profiling, decreased protein biosynthesis and reduced levels of short-lived proteins, suggesting translation inhibition (Fig. 1M). Taken together, our results suggest that PRMT9 plays an oncogenic role in AML or LSC maintenance, by promoting PABP1 methylation mediated translation activity. Further studies are needed to explore if targeting PRMT9 with newly identified lead compound ablates LSCs activity. Figure 1 Figure 1. Disclosures Marcucci: Novartis: Other: Speaker and advisory scientific board meetings; Agios: Other: Speaker and advisory scientific board meetings; Abbvie: Other: Speaker and advisory scientific board meetings.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4842-4842
Author(s):  
Q.F. Xiao ◽  
Zi X. Chen ◽  
Dan D. Liu ◽  
Jian N. Cen ◽  
Jun He ◽  
...  

Abstract The diagnosis of myelodysplastic syndrome (MDS) is made largely on the dysplastic morphology of BM cells from aspiration or biopsies. Prognosis scored by IPSS is depending on the percentage of marrow myeloblasts and the clonal cytogenetic abnormalities. To expand the understanding of genetic defects in hematopoietic cells of MDS in hope of finding novel genes correlated to pathogenesis and provide possible diagnostic marker for MDS, we have applied microarray to analyze the clinical samples from MDS patients. Total RNAs of CD34+ cells from 8 patients ( 2 RAEBt,2 RAEB,2 RA,1 RAS,1 CAA ) and one healthy people were extracted followed by a double in vitro transcription to circumvent the limited number of CD34+ cells. Following a modified Affymetrix target amplification protocol. Biotinylated cRNA was synthesized from 50 ng total RNA by double-round amplification and hybridized to an Human Genome U133 Plus 2.0 Array (Affymetrix). From the expression profile of 18404 different genes, we revealed that DNTT,MLL3,IL1R2,MAPK1,IGLL1 were down regulated while EGR-1, Rap1GAP or MAF were up regulated compared with normal controls. Most notably, Dlk1 was up regulated in MDS, while down regulated in AML and normal. By real-time RT-PCR we confirmed that in BMNCs the median levels of Dlk1 transcript in patients with RA and RAS were 2.55 (range, 0.00–23.7), RAEB and RAEBt were 8.24(range, 2.01–18.44), AML were 1.88 (range, 0.12–5.13), and other patients were 0.37(range, 0.00–1.79), respectively. The abundance of Dlk1 mRNA in MNCs from most MDS patients was markedly greater than that in the MNCs from others (P <0.05 ). Dlk1 expression in RAEB and RAEBt is markedly higher than AML (P <0.05 ) Forced expression of Dlk1 in transfected K562 cells resulted in faster growth than control cells, affected apoptosis induced by As2O3. and reduced the G2 arrested cells induced by TPA. By using the same experimental system we found that forced expression of Dlk1 can increase the mRNA levels of HES1 and p21WAF1 transcript variant 1. To elucidate the mechanisms we analyzed the levels of phosphorylated-p38 and p38 in Dlk1 transfected K562 cells treated with TPA. Dlk1 inhibited p38 phosphorylation while expression of p38 kept no change. These results support further investigation on the role of Dlk1 in abnormal hematopoiesis in MDSheterogeneous cell component. Diagnosis is currently depending on the dysplastic morphology of.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1090-1090
Author(s):  
Daniela Cilloni ◽  
Cristina Panuzzo ◽  
Francesca Messa ◽  
Francesca Arruga ◽  
Enrico Bracco ◽  
...  

Abstract The FoxO family of transcription factors is regulated by PI3K/Akt induced phosphorylation resulting in nuclear exclusion and degradation. Nuclear FoxO transcribes proapoptotic molecules and cell cycle inhibitors. In CML cells the TK activity of Bcr-Abl leads to the abnormal activation of downstream effectors including PI3K/Akt. The aim of this study was to investigate the role of FoxO3 in Bcr-Abl induced apoptotic arrest and cell growth and the effect of imatinib (IM) induced re-activation of FoxO3 activity in CML progenitor cells. BM cells were collected from 52 CML patients and 20 healthy donors. The expression level of FoxO3 was tested by RQ-PCR. The protein amount and localization was analyzed by Western blot and immunofluorescence, DNA binding activity was measured by EMSA. In addition, FoxO3 was analyzed in CML primary cells and CD34+ cells after IM incubation. Cell cycle and the expression levels of CD47, which has been demonstrated to increased during progression through the cell cycle and stem cell mobilization, was measured by FACS in CD34+ cell population. In addition K562 cells was transfected with pECE-FoxO3 to clarify FoxO3 effects on cell growth and apoptosis. Finally we used our already set up model of Drosophila melanogaster (Dm) transgenic for human Bcr-Abl to study the pathway leading to FoxO3 inactivation. We found that, despite either FoxO3 mRNA levels or protein amount are similar in CML cells compared to controls, FoxO3 protein is equally distributed in the nucleus and cytoplasm in controls but it is completely cytoplasmatic in CML cells and it enters the nucleus during in vivo IM treatment or in vitro IM incubation. Additionally, FoxO3 DNA binding activity in CML patients is completely absent at diagnosis and reappears after IM treatment. Moreover FoxO3 overexpression in transfected cells results into a 49±9 % reduction of proliferation which was further reduced of 75±5 % after IM incubation. Furthermore, we demonstrated that IM incubation results into the reactivation of FoxO3 in Ph+ CD34+ cells inducing quiescence into this population as demonstrated by the comparison of cell cycle kinetics and by a decreased expression of CD47. Finally, the progeny obtained from the crossbreeding of Bcr-Abl flies and flies transgenic for FoxO showed a rescue of FoxO phenotype demonstrating that FoxO inactivation is Bcr-Abl mediated. Overall, these in vitro and in vivo experiments suggest that FoxO3 is inactivated in CML cells and its delocalization is mainly dependant from Bcr-Abl activity. The antiproliferative activity of IM may be mediated by FoxO3 re-localization. On the other side, FoxO3 re-activation induced by IM results into a quiescence of Bcr-Abl CD34+ progenitor cells, which raises a hypothesis that FoxO3 could play a role in IM resistance. This investigation was conducted by CML Correlative Studies Network (CCSN), TOPS, which is sponsored by Novartis Oncology


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3907-3907
Author(s):  
Costanza Bogani ◽  
Paola Guglielmelli ◽  
Niccolò Bartalucci ◽  
Miguel Aracil ◽  
Maria Fe Paz ◽  
...  

Abstract Abstract 3907 Poster Board III-843 Plitidepsin (Aplidin®) is a novel cyclic depsipeptide derived from the marine tunicate Aplidium albicans, currently obtained by chemical synthesis, that is under Phase II clinical development. Plitidepsin is effective against a large panel of tumor cells, and although precise action mechanisms have still to be ascertained the drug induces an oxidative stress, activation of Rac1 GTPase and inhibition of protein phosphatases, overall leading to sustained activation of JNK and p38MAPK. In a previous report (Verrucci M et al, ASH 2008, 2787A) we evaluated Plitidepsin activity in the GATA-1low murine model of myelofibrosis. Plitidepsin corrected thrombocytopenia of myelofibrotic mice, reduced the frequency of megakaryocytes (Mk) and normalized angiogenesis in the bone marrow, and prevented extramedullary hematopoiesis. In the present study, we assessed the effects of Plitidepsin on cell lines harboring homozygous (HEL and UKE-1, a gift of W. Fiedler) or heterozygous (SET2) JAK2V617F mutation and on cells from patients (pts) with myeloproliferative neoplasms (MPN). In a short-term (3 days) proliferation assay we found that Plitidepsin prevented cell growth with IC50 values of 1.0±0.3 nM for HEL, 0.5±0.03 nM for UKE-1, and 0.8±0.02 nM for SET2, that were all lower than 1.5±0.1 nM for the BCR/ABL mutated K562 cell line (P<.001 in case of UKE-1 cells). Also Ba/F3 cells transduced with the V617F allele (a gift of R. Skoda) were found more sensitive to Plitidepsin (IC50= 0.03±0.01 nM) than the wild-type counterpart (IC50= 0.4±0.03 nM; P<0.02). Similar results were obtained using a 14-day clonogenic assay in agar cultures. These data indicated that Plitidepsin was active at very low nanomolar concentrations against cell lines harboring JAK2V617F mutation. We then evaluated the effects of Plitidepsin on the growth of BFU-E, CFU-GM and CFU-Mk from MPN pts; all five Polycythemia Vera (PV) and 4/5 Primary Myelofibrosis (PMF) pts analyzed were JAK2V617F mutated. As shown in the Table, PMF pts presented significantly lower IC50 value than controls (Ctrl; P<.002) for all type of clonogenic progenitors; cells from PMF pts resulted also significantly more sensitive to Plitidepsin than those from PV patients (P<.02), while the difference between PV and Ctrl did not reach the significance level. To evaluate whether Plitidepsin also affected the latest stages of differentiation and maturation of MKs, that is the most overtly affected cell lineage in PMF, we added Plitidepsin on day +7 of a two-stage liquid culture system initiated with CD34+ cells purified from the PB of PMF patients; the generation of CD61+ Mks was measured 5 days later by FACS analysis. However, we found that the number of CD61+ cells was no different between cultures containing or not Plitidepsin, overall suggesting that the drug mainly affected early proliferation of Mk progenitors rather than influencing their differentiation. We then performed single colony genotyping to quantify the proportion of hematopoietic colonies harboring the JAK2V617F mutation which grew in growth factor-supplemented methylcellulose cultures initiated with purified CD34+ cells from PMF patients in the presence of 1 nM Plitidepsin. Initial data in 3 pts were available; in one, the proportion of JAK2-mutated BFU-E colonies decreased from 51% to 27% while no changes were observed in the other two pts. Finally, since a correlation between levels of p27(Kip1) and the response of tumor cells to Plitidepsin has been described, we measured p27 levels in different cell lines after exposure to Plitidepsin. We observed that p27 mRNA levels increased 15-fold and 30-fold in UKE1 and HEL cells, respectively, compared to K562 cells after 24 hr with 1nM Plitidepsin; such an increase was mirrored by a protein content 1.9- to 3.5-fold greater than baseline in UKE-1 cells at 1 and 10 nM Plitidepsin, suggesting that JAK2V617F mutated cells responded to the drug by modulating their p27 levels. Collectively, we provided evidence that Plitidepsin has in-vitro activity against MPN cells, particularly from PMF pts. These results, as well as those which were previously described in the GATA1low murine model, provided the rationale for a clinical trial in patients with myelofibrosis that is being developed within the Myeloproliferative Disorders Research Consortium (MPD-MRC). Plitidepsin IC50 (nM) BFU-E CFU-GM CFU-Mk Ctrl (n=5) 8.7 ± 2.3 8.2 ± 3.5 1.7 ± 0.9 PV (n=5) 5.2 ± 2.0 7.4 ± 4.0 not done PMF (n=5) 1.1 ± 0.6 1.6 ± 0.4 0.4 ± 0.06 Disclosures: Aracil: PharmaMar: Employment. Fe Paz:PharmaMar: Employment. Vannucchi:PharmaMar: Research Funding.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3176-3176
Author(s):  
Ondrej Krejci ◽  
Mark Wunderlich ◽  
Hartmut Geiger ◽  
Fu-Sheng Chou ◽  
David Schleimer ◽  
...  

Abstract Chromosomal translocation 8;21 is present in 10–15% of patients with acute myeloid leukemia and results in the expression of the AML1-ETO (AE) fusion protein. AE expression by itself is not sufficient for malignant transformation of bone marrow progenitor/stem cells, but the nature of the cooperating genetic alterations are unknown. It is unclear whether AE facilitates acquisition of these synergizing events. We have previously shown that retroviral transduction of primary human CD34+ cells with AE causes a dramatic expansion of CD34+ progenitor cells that retain the ability to differentiate into myeloid, lymphoid, and erythroid lineages and engraft NOD/SCID mice. Consistent with mouse studies, these transgenic human cells fail to cause disease in immunodeficient mice or become transformed in vitro. These cells serve as a highly relevant pre-leukemia model for t(8;21) leukemogenesis. Here we demonstrate that AE downregulates genes involved in multiple DNA repair pathways, an effect that is evident as early as two days after retroviral transduction of human CD34+ cells. The human AE cells show delayed repair of induced DNA damage as shown by kinetics of a comet assay. During in vitro culture, AE cells show spontaneous accumulation of DNA damage, as demonstrated by γH2AX staining. Using a transgenic mutation frequency assay based on the lacZ reporter gene, we show that the in vivo mutation frequency of murine BM cells was increased approximately two fold by AE expression. Possibly as a result of this chronic DNA damage, AE cells have increased p53 protein expression and an upregulation of multiple p53 response genes. This results in increased basal apoptosis and enhanced sensitivity to DNA damaging agents compared to control cells. Intriguingly, microarray data indicate that t(8;21) patient samples also exhibit increased expression of p53 response genes when compared to other AML patient samples. Inhibition of the p53 pathway by shRNA interferes with the G1 cell cycle checkpoint and increases the resistance of AE cells to DNA damage. We thus speculate that AML1-ETO may indeed facilitate accumulation of additional genetic alterations by suppressing endogenous DNA repair, but that this activity also renders the cells more susceptible to stress. It is possible that the superior outcome of t(8;21) patients is partly due to an activated p53 pathway, and that loss of the p53 response pathway in disease progression will consequently negatively impact overall outcome. We are currently investigating whether a “p53 activation” profile can be used as a prognostic tool in AML.


Blood ◽  
2006 ◽  
Vol 107 (4) ◽  
pp. 1599-1607 ◽  
Author(s):  
Ofer Margalit ◽  
Hila Amram ◽  
Ninette Amariglio ◽  
Amos J. Simon ◽  
Sigal Shaklai ◽  
...  

The BCL6 transcriptional repressor mediates survival, proliferation, and differentiation blockade of B cells during the germinal-center reaction and is frequently misregulated in B-cell non-Hodgkin lymphoma (BNHL). The p53 tumor-suppressor gene is central to tumorigenesis. Microarray analysis identified BCL6 as a primary target of p53. The BCL6 intron 1 contains a region in which 3 types of genetic alterations are frequent in BNHL: chromosomal translocations, point mutations, and internal deletions. We therefore defined it as TMDR (translocations, mutations, and deletions region). The BCL6 gene contains a p53 response element (p53RE) residing within the TMDR. This p53RE contains a motif known to be preferentially targeted by somatic hypermutation. This p53RE is evolutionarily conserved only in primates. The p53 protein binds to this RE in vitro and in vivo. Reporter assays revealed that the BCL6 p53RE can confer p53-dependent transcriptional activation. BCL6 mRNA and protein levels increased after chemotherapy/radiotherapy in human but not in murine tissues. The increase in BCL6 mRNA levels was attenuated by the p53 inhibitor PFT-α. Thus, we define the BCL6 gene as a new p53 target, regulated through a RE frequently disrupted in BNHL.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2610-2610
Author(s):  
Camelia Iancu-Rubin ◽  
Faye Feller ◽  
David Gajzer ◽  
John Mascarenhas ◽  
Ronald Hoffman

Abstract Abstract 2610 Megakaryocytopoiesis consists of a succession of events in which MK progenitors initially proliferate and acquire lineage-specific markers, followed by polyploidization and cytoplasmic maturation. MK maturation culminates in the formation of cytoplasmic extensions (i.e. proplatelets) that leads to platelet shedding into the circulation. Panobinostat (LBH589) is a histone deacetylase inhibitor that has antiproliferative and cytotoxic effects on several types of cancer cells including blood cells from patients with hematological malignancies. One of the major adverse events associated with LBH589 treatment is thrombocytopenia. In this study, we hypothesize that the effects of LBH589 on thrombopoiesis might occur by targeting acetylation of histone and/or non-histone proteins resulting in defective platelet production. To test this hypothesis we investigated the effects of LBH589 on megakaryocytopoiesis in MK cell lines (i.e. HEL JAK2V617F positive cells) and in primary human MK. First, we tested the effects of LBH589 on the ability of human CD34+ cells to generate MK colony forming units (CFU-MKs). Neither CFU-MK or CFU-MIX derived colony formation was reduced in the presence of LBH589. To evaluate the effects of LBH589 on parameters of MK maturation, MK were generated in vitro from peripheral blood-derived CD34+ cells by employing an expansion culture system containing SCF and TPO for 6 days followed by 8 additional days incubation in the presence of TPO. These studies were pursued in the presence or absence of LBH589. Treatment with LBH589 did not significantly influence the number of CD61+ MK (i.e. control = 55.8%; 2.5nM LBH589 = 45.2%, p value=0.109; 5nM LBH589=38.5%, p value=0.095, of viable 7-AAD−/CD61+ cells) or the degree of polyploidization (i.e. control = 17.4%; 2.5nM LBH589 = 14.4%86.7, p value=0.157; 5nM LBH 589=12.8%, p value=0.116, cells with >4N DNA content). Culture-derived platelets were analyzed phenotypically and quantitated by means of dual labeling with anti-CD41 antibodies and with thiazole orange (TO) in order to identify new reticulated platelets. The percentage of CD41+/TO+ platelets derived from MK generated in the presence of LBH589 was significantly reduced (i.e. 2.5nM LBH589=11%, p value 0.046 and 5nM LBH589=9%, p value=0.011, CD41+/TO+ cells) as compared with MK generated in the absence of LBH589 (18.5% CD41+/TO+ cells). These findings were consistent with the observation of significant numbers of proplatelet-bearing MKs in control cultures but not in LBH 589-treated cultures. Collectively, these data suggest that LBH589 impairs platelet production while having a minimal effect on MK commitment, cytoplasmic maturation or polyploidization. To better understand the mechanisms responsible for such effects on thrombopoiesis, RNA extracted from control MK and from MK treated in vitro with LBH589 was analyzed by real time quantitative PCR to evaluate GATA-1 and NF-E2 expression. GATA-1 and NF-E2 mRNA levels were unchanged after treatment with LBH589. We found, however, that LBH589 induced a 4.8 to 7.5-fold increase in histone H3 acetylation. These data suggest that the negative impact of LBH589 on MK maturation was not mediated by its effects on chromatin but rather was possibly due to its effects on acetylation of nonhistone proteins. We demonstrated that LBH589 treatment increased acetylation of tubulin, a non-histone cytoplasmic protein that is a component of the microtubule (MT) cytoskeleton. The later stages of MK maturation are highly dependent on MT which represent the structural scaffold for proplatelet extension and enables the transport of cytoplasmic organelles into nascent platelets. The changes in the acetylation status of tubulin are critical for proper MT function and are mediated by HDAC6 which we found by Western blot analysis to be inhibited by LBH589 treatment. Based on these findings we suggest that LBH589-induced changes in tubulin acetylation result in aberrant MT function which in turn, leads to defective proplatelet and platelet formation. These nonhistone protein modifications might serve as a drug target for the development of novel agents (LBH589) to treat patients with extreme thrombocytosis due to underlying myeloproliferative neoplasms. Disclosures: Iancu-Rubin: Novartis: Research Funding. Hoffman:Novartis: Research Funding.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2891-2891
Author(s):  
Sampath Ramachandiran ◽  
Xiangxue Guo ◽  
Laurent Garderet ◽  
Souhila Ikhlef ◽  
Jean El-cheikh ◽  
...  

Abstract Abstract 2891 The proteasome inhibitor, bortezomib, is a powerful agent for the treatment of Multiple Myeloma (MM). However, many patients fail to achieve an optimal response. To identify biomarkers that could predict an optimal response to bortezomib, we assessed the role of the noncanonical NF-kB pathway in the response to bortezomib. Our in vitro studies identified that NF-kB2/p100 protein levels correlated directly with bortezomib sensitivity. Based on this finding, we measured at diagnosis the mRNA levels of different NF-kB2 regions in 64 patients schedule to receive bortezomib-based regimens. While no difference in expression of the 5'end regions was found between the CD138 (+) and CD138 (-) cells in these patients, half of them demonstrated low or absent 3'end expression levels in the CD138 (+) cells. Among forty-seven patients treated with four cycles of Bortezomib and dexamethasone, we found that low plasma cell NF-kB2 3'end levels were associated with a lower overall response rate (low: 47.8% vs. high: 96%, see table). In contrast, low plasma cell NF-kB2 3'end levels failed predict bortezomib response when lenalidomide or thalidome was added to the therapeutic regimen. To further understand the cause for NF-kB2 truncation, we are currently analyzing RNA sequencing in 3 non-responder patients and whole genome sequencing of 38 MM patients to determine the potential causative mechanisms. Preliminary results suggest that NF-kB2 truncation result from multiple genetic alterations, including: premature stop codons produced by ALU insertions, inversions and splicing variants. Overall, this study substantiates the basis of a more precise therapeutic algorithm according to NF-kB2 3'end plasma cell levels, which include the use of bortezomib and dexamethasone in patients with high NF-kB2 3'end plasma cell levels and immunomodulators or DNA damaging agents in patients with low NF-kB2 3'end plasma cell levels.BD+BTD or BRD++NF-kB2 3' end mRNA (CD 138(+)/CD138(−)LowHighLowHigh> Very Good Partial Response5 (21.7)9 (37.5)1 (11)4 (50)Partial Response6 (26.1)14 (58.3)7 (78)4 (50)Stable Disease4 (17.4)1 (4.2)0 (0)0 (0)Progressive disease8 (34.8)0 (0)1 (12)0 (0)Overall Response11 (47.8)23 (95.8)8 (88)8 (100)*Bortezomib + Dexamethasone**Bortezomib+Lenalidomide/Thalidomide+Dexamethasone Disclosures: Kaufman: Millenium, Onxy, Novartis, Keryx: Consultancy; Merck, Celgene: Research Funding.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 19-19
Author(s):  
Alejandro Roisman ◽  
Emmalee R. Adelman ◽  
Natalia Weich ◽  
Aristeidis G. Telonis ◽  
Dean Wade ◽  
...  

Abstract Aging causes a gradual decline in hematopoietic stem cell (HSC) function, which increases the risk for hematological malignancies. While much has been done in murine models, human HSC aging impairment is less understood. We recently showed that Krüppel-like transcription factor 6 (KLF6) is among the top downregulated genes during human HSC aging, which correlates with H3K27ac loss at several upstream putative enhancers. Moreover, loss of KLF6 in human CD34 + cells resulted in impaired in vitro differentiation, increased colony-forming potential and a transcriptional profile similar to that of aged CD34 + CD38 - cells. We hypothesized that age-acquired deregulation of KLF6 may be a key player in age-related HSC dysfunction and sought to fully characterize this. Thus, we isolated CD34 + cells from young (&lt;32 y.o) and aged (&gt;65 y.o.) healthy donors and performed CRISPR-Cas9 genome editing and transcriptional activation of KLF6, respectively, followed by epigenetic and transcriptional reprogramming, in vivo hematopoietic reconstitution, and analysis of DNA damage, apoptosis, and reactive oxygen species (ROS) levels. KLF6 knock-out (KO) and non-targeting control (NTC) cells from young healthy donors were engrafted into immunodeficient NSGS mice. Hematopoietic reconstitution analysis showed that KLF6 KO cells led to increased myeloid and reduced lymphoid reconstitution in peripheral blood (PB; p&lt;1.62 -7) and an increase in immunophenotypically defined HSC and CD34 + CD38 - progenitor fractions in the bone marrow (BM; p=0.02, and p=0.04, respectively). H3K27ac analysis of KLF6 KO cells revealed a loss of 3,390 ChIP-seq peaks (FDR &lt; 0.05) and 285 peaks gained. Functional annotation using ChIP-Enrich showed that H3K27ac loss associates with myeloid homeostasis, erythroid differentiation and oxidative stress (FDR &lt; 0.05). Three putative enhancer (E) regions upstream of the KLF6 locus showed loss of H3K27ac with aging. Depletion of the E1 but not E2 or E3 regions phenocopied in vitro and in vivo findings of KLF6 KO. Transcription factor (TF) ChIP-seq data analysis revealed FLI1, ERG, and RUNX1 binding overlapping the E1 region. Knockdown of FLI1 but not ERG or RUNX1 led to an increase in KLF6. Notably, FLI1 mRNA levels, but not ERG or RUNX1, are increased during normal aging. We next performed in vitro KLF6 activation in aged CD34 + (KLF6a) cells using a dCas9-VP64 system to test if we could rejuvenate these cells. KLF6a cells exhibited a decrease in their in vitro myeloid differentiation potential, compared to aged NTC CD34 + cells (p&lt;0.0041), and behaved instead similar to young controls. ChIP-seq analysis of KLF6a showed marked decrease of H3K4me1 (n=3,273 peaks) with relatively few regions with increased H3K4me1 (n=602) (FDR &lt; 0.05). In contrast, we observed an increase in H3K27ac (n=3,361 peaks) with only 71 peaks lost compared to aged NTC (FDR &lt; 0.05). Regions that gained H3K27ac in KLF6a were associated with platelet activation, cell junction and adhesion. In vivo analysis of KLF6a cells injected into NSGS mice revealed a significant reduction in the PB myeloid fraction compared to NTC (p&lt;1.2-8), with a concomitant expansion in the lymphoid compartment (p&lt;4.4 -11). BM composition analysis at week 16 showed a decrease in the HSC fraction in KLF6a cells (p=0.0029) as well as a reduction in CD34 +CD38 -, CD34 +CD38 + and MEPs (p=0.036, p&lt;0.0001 and p=0.041, respectively). We next examined the impact of KLF6 modulation on DNA damage and observed that young human KLF6 KO cells had a significant increase in gH2AX and 53BP1 (p&lt;0.0001, for both) whereas KLF6a in aged CD34 + cells exhibited reduced gH2AX and 53BP1 foci in comparison to aged NTC (p&lt;0.0001, for both). In addition, apoptotic levels in KLF6 KO cells were higher than in NTC cells (p=0.006) whereas aged KLF6a cells showed a reduction in the incidence of apoptotic cells compared to NTC (p=0.019). Finally, ROS analysis in young KLF6 KO showed increased levels of total and mitochondrial ROS compared to NTC (p=0.0008 and p&lt;0.0001, respectively) whereas both ROS fractions were reduced in KLF6a cells (p=0.0002 and p&lt;0.0001, respectively). In summary, these results show that the FLI1-KLF6 axis plays a key role in regulating HSPC aging and that KLF6 is required for normal HSPC function and differentiation. In addition, normalization of KLF6 levels in aged HSPCs resulted in reprogramming and rejuvenation HSPCs, confirming the central role of this TF in aging HSPC biology. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2231-2231
Author(s):  
Fabienne Isken ◽  
Björn Steffen ◽  
Birgit Markus ◽  
Claudia Hömme ◽  
David Kosel ◽  
...  

Abstract MicroRNAs (miRNAs) play an important role in differentiation, regulation of cell growth, cell death and potentially cancer pathogenesis. In the current study, we profiled expression of 154 human miRNAs by using a stem-loop reverse transcription and real-time PCR approach in 50 patients with AML and 12 control samples (normal bone marrow (NBM) samples and CD34+ cells). All of these samples were simultaneously analysed by whole genome mRNA microarrays. Out of all miRNAs tested, more than 30 were analyzed in a second, independent group of 48 AML samples and 4 controls. Five miRNAs showed differentiation associated expression changes which were confirmed by in-vitro differentiation of HL-60 cells with all-trans retinoic acid (ATRA) or 12-O-tetradecanoylphorbol-13-acetate (TPA). Overall, we found six miRNAs, including the miR-221 and 222 cluster, that showed substantially different expression between AML and CD34+ cells/ NBM. In class prediction analyses using support vector machines, 92% of the AML samples and 100% of the control samples were correctly predicted. We also looked for differentially regulated miRNAs in FAB subtypes and identified several miRNAs with significantly different expression. For example, high expression levels of miR-26a, 23a and 27b occurred in FAB M5. mRNA levels were analyzed with regard to differences in miRNA expression. Significant negative correlations with specific mRNAs were observed for several miRNAs, for example miR-23b. When mRNA expression patterns were analyzed for dichotomized miRNA levels, close associations between specific miRNAs and gene expression clusters were observed. For example, miR-10a levels were closely associated with the expression of hox genes. Taken together, the distinct expression patterns of miRNAs observed in AML strongly suggest involvement of the described miRNAs in both hematopoietic differentiation and pathogenesis of AML.


2021 ◽  
Vol 22 (19) ◽  
pp. 10814
Author(s):  
Emanuela Chiarella ◽  
Annamaria Aloisio ◽  
Stefania Scicchitano ◽  
Katia Todoerti ◽  
Emanuela G. Cosentino ◽  
...  

Leukemias derived from the MLL-AF9 rearrangement rely on dysfunctional transcriptional networks. ZNF521, a transcription co-factor implicated in the control of hematopoiesis, has been proposed to sustain leukemic transformation in collaboration with other oncogenes. Here, we demonstrate that ZNF521 mRNA levels correlate with specific genetic aberrations: in particular, the highest expression is observed in AMLs bearing MLL rearrangements, while the lowest is detected in AMLs with FLT3-ITD, NPM1, or CEBPα double mutations. In cord blood-derived CD34+ cells, enforced expression of ZNF521 provides a significant proliferative advantage and enhances MLL-AF9 effects on the induction of proliferation and the expansion of leukemic progenitor cells. Transcriptome analysis of primary CD34+ cultures displayed subsets of genes up-regulated by MLL-AF9 or ZNF521 single transgene overexpression as well as in MLL-AF9/ZNF521 combinations, at either the early or late time points of an in vitro leukemogenesis model. The silencing of ZNF521 in the MLL-AF9 + THP-1 cell line coherently results in an impairment of growth and clonogenicity, recapitulating the effects observed in primary cells. Taken together, these results underscore a role for ZNF521 in sustaining the self-renewal of the immature AML compartment, most likely through the perturbation of the gene expression landscape, which ultimately favors the expansion of MLL-AF9-transformed leukemic clones.


Sign in / Sign up

Export Citation Format

Share Document