Isolation and Functional Characterization of a Novel “Oxidative State – low” Leukemic Population with Stem Cell Properties and Potential Resistance to Chemotherapy In Acute Myeloid Leukemia.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1580-1580
Author(s):  
Eleni D. Lagadinou ◽  
Dimitra Kokkinou ◽  
Elena Siapati ◽  
George Vassilopoulos ◽  
Craig T. Jordan ◽  
...  

Abstract Abstract 1580 Most adults with acute myeloid leukemia (AML) are not cured with current treatments due to primary chemo-resistance or relapse. Emerging evidence suggests that the pool of leukemic blasts is heterogeneous and disease persistence is due to a subset of leukemic (-stem) cells able to evade chemotherapy and sustain tumor growth. Cell surface marker expression has proven to be a valuable tool to isolate and study leukemic stem cells (LSC) which, similarly to normal hematopoiesis, are shown to reside in the CD34+/CD38- leukemic fraction. However, recent data indicate that the phenotype of LSC varies from patient to patient and it seems likely that no single phenotypic signature exists to uniformly identify LSC. Besides immunophenotype, isolating LSC on the basis of functional properties unique for these cells may enforce our understanding of AML biology and provide the basis to develop more effective therapies. Reactive oxygen species (ROS) regulate essential cellular functions such as self-renewal, proliferation and apoptosis. In normal neurogenesis and hematopoiesis, ROSlow states correlate with self-renewal and ROShigh is associated with differentiation. In malignant tissues, although cancer cells are commonly more oxidized than their normal counterparts, some cancer stem cells are shown to contain low ROS levels, a feature associated with increased resistance to therapy (Nature. 2009;458:780-783). We hypothesized that LSCs also reside in a less oxidized state than bulk leukemic cells, a condition which promotes self-renewal and confers resistance to chemotherapy. To validate this hypothesis, we evaluated the redox state of leukemic blasts isolated from bone marrow or peripheral blood from 21 AML and 2 high-risk MDS patients. Loading of cells with the fluorescent probe DCF-DA showed that primary AML specimens have a broad range of oxidative state, with cells clearly falling into ROShigh and ROSlow populations (ROSlow=11.5±9%). Phenotypic analyses of AML specimens with respect to primitive cell surface markers indicated that the ROSlow gate represented 18 ± 17% of the phenotypically primitive CD34+/CD38- cells and was significantly more enriched in CD34-/CD38- leukemic cells in comparison to ROShigh. We isolated ROSlow and ROShigh leukemic subsets by flow cytometric sorting on the basis of their DCF fluorescence from 11 AML patients' samples and analyzed them for stem cell properties and drug sensitivity. Importantly, we used the differential redox state and not phenotypic markers to isolate distinct leukemic subpopulations. Morphological evaluation of sorted CD45/SS blast gated, DCFlow and DCFhigh cells demonstrated that both subpopulations were leukemic. Comparative analysis of the cell cycle distribution after staining with Ki67 and 7AAD indicated in most cases that ROSlow cells are quiescent, in contrast to ROShigh and total blast cells which are more actively cycling. Despite their predominant quiescent state, ROSlow leukemic cells were able both to grow as colonies in CFU assays and also to engraft in NOD SCID mice in pilot experiments, suggesting the existence of both leukemic “progenitor” and “stem” cell types within the ROSlow leukemic fraction. Based on these data, we challenged primary AML specimens with conventional chemotherapy agents (daunorubicin and AraC). Intriguingly, ROSlow cells preferentially survived exposure to either antileukemic agent in vitro. Taken together, our data identify a novel quiescent “oxidative state – low” leukemic population from patients with AML/MDS at diagnosis, which displays stem cell properties and exhibits functional differences related to drug sensitivity. The detailed molecular and functional characterization of this novel leukemic population is the subject of our ongoing studies. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1231-1231
Author(s):  
Andrew Lewis ◽  
Chun Shik Park ◽  
Monica Puppi ◽  
H. Daniel Lacorazza

Acute myeloid leukemia (AML) develops from sequential mutations which transform hematopoietic stem and progenitor cells (HSPCs) in the bone marrow into leukemic stem cells (LSCs) which drive the progression of frank leukemia. Especially poor outcomes in elderly patients coupled with frequent relapse have led to a dismal 28.3% 5-year survival, warranting the need for innovative therapeutic approaches. Successful targeted therapy will selectively eliminate LSCs, which possess distinct characteristics enabling self-renewal and chemotherapeutic resistance, while sparing normal HSPCs. We theorized that KLF4, a zinc finger transcription factor, maintains key self-renewal pathways in LSCs due to its known importance in preserving stemness in embryonic and cancer stem cells. KLF4 alters gene transcription through its activating and repressing domains as well as remodeling chromatin through various epigenetic mechanisms, and work from our lab has demonstrated that loss of KLF4 in leukemia driven by the BCR-ABL fusion oncogene results in depletion of LSCs (Park et. al in revision) while enhancing self-renewal of hematopoietic stem cells. To address this hypothesis, mice featuring floxed Klf4 gene (Klf4fl/fl) were crossed with transgenic Vav-iCre mice to produce mice with hematopoietic-specific deletion of Klf4 (Klf4Δ/Δ). The murine t(9;11)(p21;q23) translocation (MLL-AF9 or MA9) transduction model has previously been shown to reflect clinical disease attributes, and represents the MLL-rearranged human patient subset with particularly poor prognosis and relatively higher levels of KLF4. Lin−Sca-1+c-Kit+ (LSK) cells from Klf4fl/fl and Klf4Δ/Δ mice were transduced with retrovirus containing MA9 and GFP reporter and transplanted into lethally-irradiated wild-type (WT) mice to generate trackable Klf4fl/fl and Klf4Δ/ΔAMLs. Recipients of both MA9Klf4fl/fl and Klf4Δ/Δ cells developed a rapid expansion of leukemic cells with myeloid immunophenotype by flow cytometric analysis (CD11b+Gr-1+; 68-91%), characterized as AML with latency of approximately 44.5 days. To quantify the defect induced by loss of KLF4 in the leukemic stem cell population, we performed secondary transplant of multiple limiting-dilution cell doses of primary transformed leukemic bone marrow from moribund mice. Klf4Δ/Δ AML mice exhibited significantly improved survival in all dose-cohorts, in some cases presenting no detectable leukemic cells at completion of monitoring (225 days). Limiting dilution analysis using the ELDA online software tool demonstrated a 7-fold reduction from 1 in 513 in Klf4fl/fl to 1 in 3836 in Klf4Δ/Δ AML bone marrow cells capable of leukemic initiation function (p<0.001), a hallmark of LSCs. Using the ERCre-tamoxifen inducible deletion system, Klf4 deletion 15 days post-transplant of AML significantly improved survival of Klf4Δ/Δ mice compared to controls, demonstrating KLF4 promotes maintenance of disease. Plating of leukemic bone marrow from Klf4Δ/Δ mice in methylcellulose medium revealed a reduction in serial colony-forming ability, further supporting a defect in self-renewal. To further determine the mechanisms connected to this reduction in functional LSCs, we isolated leukemic granulocyte-macrophage progenitors (L-GMPs), a population previously reported to be highly enriched for functional LSCs and representing a comparable cellular subset in human clinical samples, from Klf4fl/fl and Klf4Δ/Δ AMLs and conducted RNA-Seq to identify potential transcriptional targets of KLF4 with therapeutic promise. Taken together, these data suggest a novel function of the stemness transcription factor KLF4 in the preservation of leukemic stem cells in AML. Whereas prior models based on KLF4 expression in human cell lines and bulk AML samples have proposed a tumor suppressive role, our work suggests KLF4 supports expansion of leukemic cells with a stem cell phenotype and serial assays suggest an effect on LSC self-renewal. Further studies are being conducted to define the transcriptional and epigenetic mechanisms governing these findings. Understanding the molecular changes induced by loss of KLF4 presents promise for development of new therapies selectively targeting LSCs. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 91-91
Author(s):  
Nicolas Goardon ◽  
Emmanuele Marchi ◽  
Lynn Quek ◽  
Anna Schuh ◽  
Petter Woll ◽  
...  

Abstract Abstract 91 In normal and leukemic hemopoiesis, stem cells differentiate through intermediate progenitors into terminal cells. In human Acute Myeloid Leukemia (AML), there is uncertainty about: (i) whether there is more than one leukemic stem cell (LSC) population in any one individual patient; (ii) how homogeneous AML LSCs populations are at a molecular and cellular level and (iii) the relationship between AML LSCs and normal stem/progenitor populations. Answers to these questions will clarify the molecular pathways important in the stepwise transformation of normal HSCs/progenitors. We have studied 82 primary human CD34+ AML samples (spanning a range of FAB subtypes, cytogenetic categories and FLT3 and NPM1 mutation states) and 8 age-matched control marrow samples. In ∼80% of AML cases, two expanded populations with hemopoietic progenitor immunophenotype coexist in most patients. One population is CD34+CD38-CD90-CD45RA+ (CD38-CD45RA+) and the other CD34+CD38+CD110-CD45RA+ (GMP-like). Both populations from 7/8 patients have leukemic stem cell (LSC) activity in primary and secondary xenograft assays with no LSC activity in CD34- compartment. The two CD34+ LSC populations are hierarchically ordered, with CD38-CD45RA+ LSC giving rise to CD38+CD45RA+ LSC in vivo and in vitro. Limit dilution analysis shows that CD38-CD45RA+LSCs are more potent by 8–10 fold. From 18 patients, we isolated both CD38-CD45RA+ and GMP-like LSC populations. Global mRNA expression profiles of FACS-sorted CD38-CD45RA+ and GMP-like populations from the same patient allowed comparison of the two populations within each patient (negating the effect of genetic/epigenetic changes between patients). Using a paired t-test, 748 genes were differentially expressed between CD38-CD45RA+ and GMP-like LSCs and separated the two populations in most patients in 3D PCA. This was confirmed by independent quantitative measures of difference in gene expression using a non-parametric rank product analysis with a false discovery rate of 0.01. Thus, the two AML LSC populations are molecularly distinct. We then compared LSC profiles with those from 4 different adult marrow normal stem/progenitor cells to identify the normal stem/progenitor cell populations which the two AML LSC populations are most similar to at a molecular level. We first obtained a 2626 gene set by ANOVA, that maximally distinguished normal stem and progenitor populations. Next, the expression profiles of 22 CD38-CD45RA+ and 21 GMP-like AML LSC populations were distributed by 3D PCA using this ANOVA gene set. This showed that AML LSCs were most closely related to their normal counterpart progenitor population and not normal HSC. This data was confirmed quantitatively by a classifier analysis and hierarchical clustering. Taken together, the two LSC populations are hierarchically ordered, molecularly distinct and their gene expression profiles do not map most closely to normal HSCs but rather to their counterpart normal progenitor populations. Finally, as global expression profiles of CD38-CD45RA+ AML LSC resemble normal CD38-CD45RA+ cells, we defined the functional potential of these normal cells. This had not been previously determined. Using colony and limiting dilution liquid culture assays, we showed that single normal CD38-CD45RA+ cells have granulocyte and macrophage (GM), lymphoid (T and B cell) but not megakaryocyte-erythroid (MK-E) potential. Furthermore, gene expression studies on 10 cells showed that CD38-CD45RA+ cells express lymphoid and GM but not Mk-E genes. Taken together, normal CD38-CD45RA+ cells are most similar to mouse lymphoid primed multi-potential progenitor cells (LMPP) cells and distinct from the recently identified human Macrophage Lymphoid progenitor (MLP) population. In summary, for the first time, we show the co-existence of LMPP-like and GMP-like LSCs in CD34+ AML. Thus, CD34+ AML is a progenitor disease where LSCs have acquired abnormal self-renewal potential (Figure 1). Going forward, this work provides a platform for determining pathological LSCs self-renewal and tracking LSCs post treatment, both of which will impact on leukemia biology and therapy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2560-2560
Author(s):  
Larissa Balaian ◽  
Anil Sadarangani ◽  
George F. Widhopf ◽  
Rui-kun Zhong ◽  
Charles Prussak ◽  
...  

Abstract Abstract 2560 The mammalian orphan receptor tyrosine kinase-1 (ROR1) is expressed in a wide-variety of tissues during early embryonic development. By the late stages of embryogenesis the expression of this developmentally important protein is greatly diminished. Although not expressed in the tissues of post-partum animals, the ROR1 protein is expressed on neoplastic cells in chronic lymphocytic leukemia (CLL), some B-cell malignancies, and a variety of different carcinomas. We examined for expression of ROR1 in primary acute myeloid leukemia (AML) cells harvested from marrow aspirates and their normal counterparts by whole transcriptome paired-end RNA sequencing and by flow-cytometric analyses. These studies revealed selective expression of ROR1 in 62 (35%) of 179 AML samples examined. Many of these samples were found to have cells that co-expressed ROR1 and CD34, suggesting that ROR1 was present on the self-renewing leukemia stem-cell population, which resides in the marrow niche and potentially accounts for resistance to many cytotoxic drugs used in therapy. We investigated the activity of a chimeric anti-ROR1 mAb found effective in clearing CLL cells (UC99961) on AML expansion, growth, and renewal in a leukemia-stem-cell supportive niche assay. Mouse marrow cells lines SL/SL and M2–10B4 (transfected to produce hSCF,hIL3 and hIL3, hG-CSF respectively) were mixed 1:1 after mitomycin-C treatment, and used as a SLM2 stromal monolayer. CD34+ cells were selected from ROR1-positive (n=6) or negative (n=4) AML primary samples. As a normal control, CD34+ cells from cord blood (CB) were used (CB, n=3). In some experiments CD34+ cells were transfected with a GLP-lentivirus prior to co-culture. At the initiation of the co-culture, 10–50 μg/ml of the chimeric anti-ROR-1 mAb (UC99961) or control hIgG were added to the cultures. Two weeks after co-culture initiation, both stromal attached and floating cells were collected and their survival investigated by colony forming assay in methylcellulose. The UC99961 mAb was not cytotoxic to CB or ROR1-negative AML samples. In contrast, the UC99961 mAb provided a dose-dependent inhibition of colony formation for all ROR-1-positive AML samples examined. These results demonstrate the in vitro anti-leukemic specificity of this anti-ROR1 mAb in down-regulating AML stem and progenitor cell populations, without effecting normal CD34+ stem cells. To analyze the effect of ROR1 ligation on AML stem cell populations exclusively, AML self-renewal assays (2-ry colonies) were performed. In these studies, ROR1–positive AML samples were divided based on their response to mAb treatment. Half of the samples (n=3; 50%) demonstrated statistically significant (up to 90%) dose-dependent decreases in colony formation. However, another half was non-responsive and no correlation was found between ROR1 expression on leukemia CD34+ cells and response to anti-ROR1 mAb treatment in the self-renewal assays. Again UC99961 mAb treatment did not negatively impact CD34+ cells from CB or ROR1-negative AML, confirming the specificity and selective toxicity of the mAb for ROR1+ AML stem cells. These studies reveal selective expression of ROR1 on leukemia-stem-cells of large subset of AML patients. Furthermore, this study demonstrates that an anti-ROR1 mAb (UC99961) can inhibit survival and self-renewal in LSC supportive niche assays. Targeted ROR1 inhibition may represent a vital component of therapeutic strategies aimed at eradicating therapeutically recalcitrant malignant stem cells in AML and potentially other refractory cancer-stem-cell-driven malignancies. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A465-A465
Author(s):  
Catherine Sabatos-Peyton ◽  
Tyler Longmire ◽  
Lisa Baker ◽  
Nidhi Patel ◽  
Anne-Sophie Wavreille ◽  
...  

BackgroundTIM-3 is expressed on leukemic stem cells (LSCs) and blasts in AML,1 2 and TIM-3 expression on MDS blasts correlates with disease progression.3 Functional evidence for TIM-3 in AML was established with an anti-TIM-3 antibody which inhibited engraftment and development of human AML in immuno-deficient murine hosts.1 TIM-3 promotes an autocrine stimulatory loop via the TIM-3/Galectin-9 interaction, supporting LSC self-renewal.4 In addition to its cell-autonomous role on LSCs/blasts, TIM-3 also has a critical role in immune system regulation, in adaptive (CD4+ and CD8+ T effector cells, regulatory T cells) and innate (macrophages, dendritic cells, NK cells) immune responses.5 MBG453 is a high-affinity, humanized anti-TIM-3 IgG4 antibody (Ab) (stabilized hinge, S228P), which blocks the binding of TIM-3 to phosphatidylserine (PtdSer). Recent results from a multi-center, open label phase Ib dose-escalation study (NCT03066648) in patients with high-risk MDS and no prior hypomethylating agent therapy evaluating MBG453 in combination with decitabine demonstrated encouraging preliminary efficacy with an overall response rate of 58%,6 and MBG453 combined with azacitidine also showed encouraging response rates.7 Preclinical experiments were undertaken to define the mechanism of action of the hypomethylating agent and anti-TIM-3 combination.MethodsTHP-1 cells (a human monocytic AML cell line) were pre-treated with decitabine and co-cultured with anti-CD3 activated healthy human donor peripheral blood mononuclear cells (PBMCs) in an Incucyte-based assay to measure cell killing. The ability of MBG453 to mediate antibody-dependent cellular phagocytosis (ADCP) was measured by determining the phagocytic uptake of an engineered TIM-3-overexpressing Raji cell line in the presence of MBG453 by phorbol 12-myristate 13-acetate (PMA)-activated THP-1 cells. Patient-derived AML xenograft studies were undertaken in immune-deficient murine hosts to evaluate the combination of decitabine and MBG453.ResultsMBG453 was determined to partially block the TIM-3/Galectin-9 interaction in a plate-based MSD (Meso Scale Discovery) assay, supported by a crystal structure of human TIM-3.8 Pre-treatment of THP-1 cells with decitabine enhanced sensitivity to immune-mediated killing in the presence of MBG453. MBG453 was determined to mediate modest ADCP, relative to controls. MBG453 did not enhance the anti-leukemic activity of decitabine in patient-derived xenograft studies in immuno-deficient hosts.ConclusionsTaken together, these results support both direct anti-leukemic effects and immune-mediated modulation by MBG453. Further studies are ongoing to determine: (1) whether MBG453 can mediate physiologically relevant ADCP of TIM-3-expressing leukemic cells; and (2) the potential of MBG453 to impact the autocrine feedback loop of TIM-3/Galectin-9.Ethics ApprovalThe human tissue used in these studies was under the Novartis Institutes of BioMedical Research Ethics Board IRB, Approval Number 201252867.ReferencesKikushige Y, Shima T, Takayanagi S, et al. TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell 2010;7(6):708–717.Jan M, Chao MP, Cha AC, et al. Prospective separation of normal and leukemic stem cells based on differential expression of TIM3, a human acute myeloid leukemia stem cell marker. Proc Natl Acad Sci USA 2011; 108(12): 5009–5014.Asayama T, Tamura H, Ishibashi M, et al. Functional expression of Tim-3 on blasts and clinical impact of its ligand galectin-9 in myelodysplastic syndromes. Oncotarget 2017;8(51): 88904–88917.Kikushige Y, Miyamoto T, Yuda J, et al. A TIM-3/Gal-9 autocrine stimulatory loop drives self-renewal of human myeloid leukemia stem cells and leukemic progression. Cell Stem Cell 2015; 17(3):341–352.Acharya N, Sabatos-Peyton C, Anderson AC. Tim-3 finds its place in the cancer immunotherapy landscape. J Immunother Cancer 2020; 8(1):e000911.Borate U, Esteve J, Porkka K, et al. Phase Ib Study of the Anti-TIM-3 Antibody MBG453 in combination with decitabine in patients with high-risk myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Blood 2019;134 (Supplement_1):570.Borate U, Esteve J, Porkka K, et al. Abstract S185: Anti-TIM-3 antibody MBG453 in combination with hypomethylating agents (HMAs) in patients (pts) with high-risk myelodysplastic syndrome (HR-MDS) and acute myeloid leukemia (AML): a Phase 1 study. EHA 2020.Sabatos-Peyton C. MBG453: A high affinity, ligand-blocking anti-TIM-3 monoclonal Ab. AACR 2016.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1608-1608
Author(s):  
Xiaomin Zheng ◽  
Tim Beissert ◽  
Brigitte Ruster ◽  
Dieter Hoelzer ◽  
Reinhard Henschler ◽  
...  

Abstract The pathogenesis of acute myeloid leukemia (AML) is strictly related to a block of terminal differentiation combined with an increased proliferation of hematopoietic progenitors (HP) in the bone marrow (BM). Furthermore an aberrant self-renewal of leukemic stem cells seems to be obligatory for the establishment of the leukemic clone in the BM. The block of differentiation is due to a deregulated function of differentiation-relevant transcription factors, whereas the proliferation is induced by aberrantly activated signaling pathways related to growth factor-dependent receptor kinases such as Flt3 or c-Kit (CD117). The aberrant self renewal can be attributed to specific pathways such as the Wnt-signaling, which play an important role also in the maintenance of the normal hematopoietic stem cell (HSC). The acute promyelocytic leukemia (APL) is characterized by the t(15;17) which leads to the expression of the PML/RAR fusion protein in the leukemic cells. PML/RAR mediates the APL phenotype given by a differentiation block at the promyelocytic level and an increased self renewal of the APL stem cells by the activation of the Wnt-signaling. The differentiation block, but not the aberrant self renewal, can be overcome by treatment with ATRA resulting ina high percentage of complete remissions. Nevertheless, if ATRA is used as a monotherapy a relapse is inevitable. APL blasts are frequently positive for constitutive activating Flt3 mutations and are constantly c-Kit-positive. Given the fact that c-Kit is a stem cell marker, the expression of c-Kit has to be considered aberrant and not related to the differentiation stage of the promyelocytes in APL. Therefore we investigated first the relationship between PML/RAR and the aberrant expression of c-Kit and then the role of aberrantly activated c-Kit for the pathogenesis of APL by studying its influence on the biology of early HSC. Here we report that i.) in contrast to the t(8;21)-associated AML-1/ETO, PML/RAR activated the c-Kit promotor in HP; ii.) the inhibition of the endogenous c-Kit kinase activity by imatinib or by AZD2171 abrogated the aberrant “replating efficiency“ of PML/RAR-positive HSC; iii.) activated c-Kit (c-Kit-D814H) accelerated cell cycle progression of Sca1+/lin- HSC; iv.) activated c-Kit blocked the differentiation of Sca1+/lin- HSC and increased their “replating efficiency“; v.) “colony forming unit-spleen“ (CFU-S) as well as “competitive repopulation“ assays“ (CRA) revealed that c-Kit-D814H strongly increased the “self renewal“-potential of Sca1+/lin- HSC; vi) c-Kit-D814H augmented the migration of Sca1+/lin- HSC into a 3D stromal spheroid model based on M2-10B4 cells, but did not have any influence on their adhesion (flow chamber on TNFalpha-stimulated HUVEC cells) as well as on their chemotaxis (SDF-1 gradient in transwell assays); vii.) c-Kit-D814H further increased the aberrant replating efficiency of PML/RAR- as well as of AML-1/ETO-positive HSC. Taken together our results strongly indicate not only that c-Kit importantly contributes to the leukemogenesis of APL, but that PML/RAR has a dual effect on c-Kit - the amplification of its expression at the promotor level as well as driving its expression in cells which normally do not have this proliferation stimulus. Thus it seems that there is a sort of positive feedback between PML/RAR and c-Kit which establishes c-Kit as a valuable therapeutic target for the treatment of APL-patients.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1202-1202
Author(s):  
Tina M. Schnoeder ◽  
Patricia Arreba-Tutusaus ◽  
Juliane Mohr ◽  
Soenke Weinert ◽  
Stephanie Frey ◽  
...  

Abstract Several genes and signaling pathways control the fine balance between self-renewal and differentiation in hematopoietic stem cells and potentially also in leukemic stem cells (LSC). Phospholipase C family members are key mediators of calcium signaling which play an important role in differentiation and proliferation of immune cells but also contribute to malignant transformation and tumorigenesis. Plcg1 is highly expressed in hematopoietic stem- and progenitor cells and also in myeloid leukemia. Plcg1 gets activated by cell extrinsic receptor stimulation and integrates signals from the cell surface. Its influence on proliferation and differentiation of hematopoietic cells may be largely independent of other bone fide mediators of self-renewal and stem cell viability such as STAT-, MEK-ERK or AKT-signaling. To which extent Plcg1-dependent signal integration is required for function and maintenance of leukemic stem cells remained so far elusive. Genetic inactivation of Plcg1 by RNAi in human AML cell lines led to decreased proliferative capacity. Likewise, knockdown of Plcg1 in AML1-ETO (AML1ETO9a) transformed murine LSK-cells resulted in reduced colony formation and decreased re-plating capacity. In order to validate these findings and to investigate the impact of Plcg1 on myeloid leukemia stem cell function, we generated a conditional knockout mouse model for Plcg1 with Exons 3-5 being flanked with loxP sites. Excision of the respective sequence by activation of a Cre-recombinase resulted in complete loss of a functional protein and transcript. LSK-cells from Plcg1f/f and Plcg1+/+ littermate controls were retrovirally infected with two different oncogenes: either MLL-AF9 (MA9-GFP) or AML1-ETO9a in combination with KRAS (AE9a/KRAS-GFP). Primary recipient mice were injected with GFP+ LSK-cells and monitored for disease development. GFP+ Kit+ cells were isolated from leukemic mice and transduced with a Cre-recombinase, followed by plating in methylcellulose. Inactivation of Plcg1 in AE9a/KRAS transformed cells significantly reduced the number of colonies and decreased re-plating capacity to three rounds. Loss of Pclg1 in MA9 transformed LSC resulted in decreased colony numbers and colony size, however, re-plating capacity was not affected to a major extent. To assess for the requirement of Plcg1 in maintenance of fully developed leukemia, we injected equal numbers of GFP+ Kit+ cells (Plcg1-/- or Plcg1+/+) into sublethally irradiated secondary recipients. Inactivation of Plcg1 was highly deleterious for AE9a/KRAS induced AML-LSC and reduced disease penetrance by more than 85%. Depletion of Plcg1 in MA9 transformed cells delayed AML development and significantly prolonged survival of recipient mice. Leukemias that developed from Plcg1-/- donors showed complete excision of Plcg1, indicating, that Plcg1 deficient leukemia can develop in an MLL-AF9 driven background. However, when transplanting MA9 transformed Plcg1-/- or Plcg1+/+ bone marrow cells into tertiary recipient mice, loss of Plcg1 significantly delayed disease progression and reduced disease penetrance. To quantify this loss of leukemic stem cells, we performed limiting dilution analysis using purified LSCs from diseased Plcg1-/- or Plcg1+/+ MA9 secondary recipient mice. LSC frequency was markedly reduced in tertiary recipients of Plcg1-depleted LSCs (1 in 78,000 Plcg1-/- vs. 1 in 3,000 Plcg1+/+). Genetic inactivation of Plcg1 in LSCs derived from primary recipient mice (either MA9 or AE9a/KRAS driven AML) led to induction of differentiation as assessed by cell morphology and immunophenotyping, and this effect was more pronounced in AE9a/KRAS transformed cells. To investigate whether transcriptional effectors of Plcg1 signaling affect the fine balance between self-renewal in MA9- and AE9a/KRAS-driven leukemia, we performed whole transcriptome analysis (RNAseq) on sorted LSCs. Ongoing analyses address the functional difference between AML-ETO and MLL-AF9 driven disease and elucidate on distinct patterns of activated gene sets depending on the oncogenic background. Taken together, Plcg1 is required for maintenance of myeloid leukemia stem cells. Understanding of its relevance in LSC biology and function may offer the opportunity to develop this relevant signaling node as a target structure in AML. Disclosures No relevant conflicts of interest to declare.


Hematology ◽  
2008 ◽  
Vol 2008 (1) ◽  
pp. 436-442 ◽  
Author(s):  
Catriona H. Jamieson

Abstract Chronic myeloid leukemia (CML) is typified by robust marrow and extramedullary myeloid cell production. In the absence of therapy or sometimes despite it, CML has a propensity to progress from a relatively well tolerated chronic phase to an almost uniformly fatal blast crisis phase. The discovery of the Philadelphia chromosome followed by identification of its BCR-ABL fusion gene product and the resultant constitutively active P210 BCR-ABL tyrosine kinase, prompted the unraveling of the molecular pathogenesis of CML. Ground-breaking research demonstrating that BCR-ABL was necessary and sufficient to initiate chronic phase CML provided the rationale for targeted therapy. However, regardless of greatly reduced mortality rates with BCR-ABL targeted therapy, most patients harbor quiescent CML stem cells that may be a reservoir for disease progression to blast crisis. While the hematopoietic stem cell (HSC) origin of CML was first suggested over 30 years ago, only recently have the HSC and progenitor cell–specific effects of the molecular mutations that drive CML been investigated. This has provided the impetus for investigating the genetic and epigenetic events governing HSC and progenitor cell resistance to therapy and their role in disease progression. Accumulating evidence suggests that the acquired BCR-ABL mutation initiates chronic phase CML and results in aberrant stem cell differentiation and survival. This eventually leads to the production of an expanded progenitor population that aberrantly acquires self-renewal capacity resulting in leukemia stem cell (LSC) generation and blast crisis transformation. Therapeutic recalcitrance of blast crisis CML provides the rationale for targeting the molecular pathways that drive aberrant progenitor differentiation, survival and self-renewal earlier in disease before LSC predominate.


2019 ◽  
Vol 20 (23) ◽  
pp. 5826 ◽  
Author(s):  
Tobias Gluexam ◽  
Alexander M. Grandits ◽  
Angela Schlerka ◽  
Chi Huu Nguyen ◽  
Julia Etzler ◽  
...  

The neuropeptide CGRP, acting through the G-protein coupled receptor CALCRL and its coreceptor RAMP1, plays a key role in migraines, which has led to the clinical development of several inhibitory compounds. Recently, high CALCRL expression has been shown to be associated with a poor prognosis in acute myeloid leukemia (AML). We investigate, therefore, the functional role of the CGRP-CALCRL axis in AML. To this end, in silico analyses, human AML cell lines, primary patient samples, and a C57BL/6-based mouse model of AML are used. We find that CALCRL is up-regulated at relapse of AML, in leukemic stem cells (LSCs) versus bulk leukemic cells, and in LSCs versus normal hematopoietic stem cells. CGRP protects receptor-positive AML cell lines and primary AML samples from apoptosis induced by cytostatic drugs used in AML therapy, and this effect is inhibited by specific antagonists. Furthermore, the CGRP antagonist olcegepant increases differentiation and reduces the leukemic burden as well as key stem cell properties in a mouse model of AML. These data provide a basis for further investigations into a possible role of CGRP-CALCRL inhibition in the therapy of AML.


Blood ◽  
2019 ◽  
Vol 134 (22) ◽  
pp. 1960-1972 ◽  
Author(s):  
Chun Shik Park ◽  
Andrew H. Lewis ◽  
Taylor J. Chen ◽  
Cory S. Bridges ◽  
Ye Shen ◽  
...  

Park et al describe a novel KLF4-mediated pathway that promotes chromic myeloid leukemia (CML) stem cell (LSC) survival. Deletion of KLF4 in a mouse model of CML decreases LSC survival through repression of Dyrk2, resulting in c-Myc depletion and increased p53 activity.


2010 ◽  
Vol 207 (4) ◽  
pp. 677-680 ◽  
Author(s):  
Omar Abdel-Wahab ◽  
Ross L. Levine

Acute leukemias are clonal disorders of hematopoiesis wherein a leukemic stem cell (LSC) acquires mutations that confer the capacity for unlimited self-renewal, impaired hematopoietic differentiation, and enhanced proliferation to the leukemic clone. Many recent advances in understanding the biology of leukemia have come from studies defining specific genetic and epigenetic abnormalities in leukemic cells. Three recent articles, however, further our understanding of leukemia biology by elucidating specific abnormalities in metabolic pathways in leukemic hematopoiesis. These studies potentially converge on the concept that modulation of reactive oxygen species (ROS) abundance may influence the pathogenesis and treatment of acute myeloid leukemia (AML).


Sign in / Sign up

Export Citation Format

Share Document