BCR/ABL Can Promote CD19+ Cell Growth but Not Render Them Long-Term Stemness

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2709-2709
Author(s):  
Donghe Li ◽  
Xuemei Zhao ◽  
Bo Jiao ◽  
Ping Liu ◽  
Ruibao Ren

Abstract Cancer stem cells are a subpopulation of malignant cells that have the capacity of both self-renewal and reconstitution of the cancer. Eradication of cancer stem cells is crucial for curing the malignant disease. Previous studies in hematopoietic malignancies showed that leukemia stem cells (LSCs) in chronic myelogenous leukemia (CML) chronic phase are originated from a hematopoietic stem cell (HSC), while LSCs in acute myeloid leukemia (AML) can either be derived from HSCs or be transformed from myeloid progenitors. But in acute B-lymphoblastic leukemia (B-ALL), the origin of leukemia stem cells is not clear. In this study, we tested whether BCR/ABL could transform B-lineage committed CD19+ cells to LSCs. We found that transducing BCR/ABL in CD19+ cells can promote their colony formation in vitro and induce B-ALL like disease in vivo. However, only BCR/ABL transduced whole bone marrow cells, but not CD19+ cells, can be transplanted multiple times in recipient mice, and the frequency of long-term LSCs from the latter ranges from 1/135 to 1/629. These studies suggest that LSCs in BCR/ABL+ B-ALL may originate from CD19- hematopoietic stem/progenitor cells and that CD19 chimeric antigen receptor (CAR) modified T cell therapy may not be effective in eradicating LSCs in BCR/ABL+ B-ALL. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 57-57
Author(s):  
Vincenzo Giambra ◽  
Catherine E Jenkins ◽  
Sonya H Lam ◽  
Catherine Hoofd ◽  
Miriam Belmonte ◽  
...  

Abstract Prior work has shown that NOTCH1 is a prominent oncogene in T-cell acute lymphoblastic leukemia (T-ALL) with activating NOTCH1 mutations occurring in over 50% of cases (Weng et al, Science 2004) and loss-of-function mutations in its negative regulator FBXW7 occurring in 8-15% of cases (O’Neil et al, J Exp Med 2007; Thompson et al, J Exp Med 2007). Subsequent work has shown that continued Notch signaling is required for maintenance of T-ALL leukemia stem cells (Armstrong et al, Blood 2009; Tatarek et al, Blood 2011; Giambra et al, Nat Med 2012). Several lines of evidence have substantiated genetic interactions between the Notch and Wnt signaling pathways in various contexts, and Wnt signaling has been shown to play important roles in hematopoietic stem cell biology and also in hematopoietic cancers such as acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Luis et al, Leukemia 2012). To address what role if any Wnt signaling may play in T-ALL, we generated primary murine leukemias by viral transduction of bone marrow progenitors with activated NOTCH1, then delivered a fluorescent Wnt reporter construct (7TGP; Fuerer & Nusse, PLoS ONE 2010) by lentiviral transduction, and retransplanted the leukemias to interrogate Wnt signaling activity in vivo. We report here that active Wnt signaling is restricted to minor subpopulations within bulk T-ALL tumors, and that these Wnt-active subsets are highly enriched for leukemia-initiating cell (LIC) activity. Moreover, using Ctnnb1loxP/loxP animals we show that inducible Cre-mediated deletion of β-catenin or enforced expression of a dominant-negative TCF construct severely compromises LIC activity. We also show that β-catenin levels are upregulated by hypoxia through Hif1a stabilization, and that deletion of Hif1a also severely compromises LIC activity. Interestingly, Wnt-active subsets are distributed diffusely throughout the marrow interstitial space suggesting that tumor infiltration induces formation of local hypoxic niches as opposed to taking up residence in pre-existing anatomic compartments with low oxygen tensions. Taken together, these results suggest a model in which hypoxic niches in vivo facilitate Hif1a-dependent accumulation of β-catenin which drives Wnt signaling and self-renewal of leukemia stem cells. Finally, we show using patient-derived xenografts that antagonism of Hif1a or Wnt signaling also compromises human LIC activity, suggesting that pharmacologic targeting of these pathways could have therapeutic application in patients with T-ALL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3584-3584
Author(s):  
Takahiro Shima ◽  
Yoshikane Kikushige ◽  
Toshihiro Miyamoto ◽  
Koichi Akashi

Abstract Abstract 3584 The 8;21 translocation, one of the most general chromosomal abnormalities in acute myelogenous leukemia (AML), encodes the AML1-ETO chimeric fusion gene. Because AML1-ETO can inhibit the CBF complex to transactivate myeloid-lineage genes in a dominant negative fashion, the high level expression of this gene plays a critical role in inhibiting differentiation of target cells, which leads to progression of AML. We, however, have reported that patients maintaining a long-term remission retain AML1-ETO expression at a very low level that can be detected by nested RT-PCR. The AML1-ETO transcripts in these patients were derived from a small fraction of t(8;21)+ hematopoietic stem cells (HSCs) capable of multilineage differentiation (PNAS 2000). In fact, previous data shown that AML1/ETO knock-in or AML1/ETO transgenic mice did not develop AML. These data suggest that acquisition of the AML1-ETO fusion is not sufficient to develop t(8;21) AML. Since t(8;21) AML cells frequently possess constitutive active mutation of c-Kit, we hypothesized that the c-Kit mutation may work as a second oncogenic hit in t(8;21)+ HSCs to transform into AML. To test the hypothesis, we extensively analyzed the existence of c-Kit mutation within AML1-ETO+ HSCs from patients maintaining remission for a long-term. CD34+CD38− HSCs were purified from the bone marrow of patients in long-term remission, and were cultured in vitro to form colonies. These HSC-derived colonies were picked up, and tested for the presence ofAML1-ETO and c-Kit mutation. Five t(8;21) AML patients with c-Kit mutation were enrolled in this study. All of 1020 blastic colonies at diagnosis were positive for both AML1-ETO and c-Kit mutation. In 7187 colonies formed in the culture of remission marrow, almost 1% (89 colonies) of these colonies expressed AML1-ETO. Surprisingly, none of these colonies possessed c-Kit mutation, indicating that AML1-ETO+ clones in remission are not identical to these in t(8;21) AML. Accordingly, it is highly likely that HSCs first acquire AML1-ETO, and a fraction of these cells additionally mutated c-Kit, resulting in transformation into AML stem cells. This is the first clear-cut evidence that human HSCs transform into AML via multi-step oncogenesis in vivo. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5257-5257
Author(s):  
Wenbin Zhong ◽  
Vesa Olkkonen ◽  
Xu Bing ◽  
Biying Zhu ◽  
Guoping Li ◽  
...  

Abstract Acute myelogenous leukemia (AML) is one of the deadliest hematological malignancies and there is at present no efficient strategy to defeat it. New detailed insight into AML leukemia stem cells (LSCs) survival will facilitate the identification of targets for the development of new therapeutic approaches. Recent work has provided evidence that LSCs are defective in their ability to employ glycolysis, but are highly reliant on oxidative phosphorylation, and the maintenance of mitochondrial function is essential for LSCs survival. It is increasingly clear that Ca2+ released constitutively from endoplasmic reticulum (ER) is taken up by mitochondria to sustain optimal bioenergetics and cell survival. Here we report three striking findings: 1) oxysterol-binding protein (OSBP)-related protein 4 (ORP4L) is expressed in LSCs but not in normal hematopoietic stem cells (HSCs). 2) ORP4L is essential for LSC bioenergetics; It forms a complex with PLCβ3 and IP3 receptor 1 (ITPR1) to control Ca2+ release from the ER and subsequent cytosolic and mitochondrial parallel Ca2+ spike oscillations that sustain pyruvate dehydrogenase (PDH) activation and oxidative phosphorylation. 3) ORP4L inhibition eradicates LSCs in vitro and in vivo through impairment of Ca2+-dependent bioenergetics. These results suggest a novel role of ORP4L in governing Ca2+ release to sustain mitochondrial function and survival of LSCs and identify ORP4L as a putative new oncoprotein and therapeutic target for LSCs elimination. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 858-858
Author(s):  
Bin Zhang ◽  
Yin Wei Ho ◽  
Sung-UK Lee ◽  
Takahiro Maeda ◽  
Claudia Huettner ◽  
...  

Abstract Abstract 858 In normal hematopoiesis, only a small population of lin-Sca-1+c-kit+ (LSK) cells with Flt3-CD150+48− immunophenotype has long-term hematopoietic stem cell (LT-HSC) capacity, whereas Flt3-CD150+CD48+ and Flt3-CD150-CD48+ LSK cells represent more differentiated multipotent progenitors (MPP1 and MPP2) without long-term engrafting capacity. Despite extensive investigation into BCR-ABL induced leukemogenesis, the impact of BCR-ABL expression on LSK subpopulations and the specific subpopulation with leukemia-initiating capacity remain unknown. Targeted expression of the BCR-ABL gene in murine hematopoietic stem and progenitor cells (HSPC), using a Tet-regulated SCL promoter, results in development of a chronic phase CML-like disorder (Blood 105:.324, 2005). Mice consistently develop leukocytosis, splenomegaly and expansion of bone marrow (BM) myeloid progenitors and primitive LSK cells following induction of BCR-ABL expression. Here we employed the SCL-tTA-BCR/ABL mouse model to investigate the effect of BCR-ABL expression on HSPC populations. BCR/ABL expression resulted in a 3-fold increase in granulocyte-macrophage progenitors (GMP) and a 1.5-fold increase in LSK cell numbers compared with non-induced controls, whereas numbers of common myeloid progenitors (CMP) and megakaryocyte-erythrocyte progenitors (MEP) were reduced. Despite expansion of total LSK cells, the number of LT-HSC was markedly reduced in the BM of BCR-ABL expressing mice (610±246 vs. 4,038±982). In contrast, an increase in MPP1 (6,150±1,813 vs. 3,185±1,247) and MPP2 (39,580±14,079 vs. 25,115±7,090) was seen. BCR-ABL mRNA expression was confirmed in each population by RT-PCR, with highest levels of expression seen in MPP cells. In vivo EdU labeling demonstrated increased cycling of LSK cells from BCR/ABL expressing mice compared to controls. We observed a vast increase in the number of GMP (11-fold), CMP (10-fold), MPP (4.5 fold) and LT-HSC (2.5 fold) in the spleen of BCR/ABL mice compared to controls. Since the functional potential of HSPC cannot be determined solely on the basis of cell surface markers, we also studied the ability of transplanted populations to generate leukemia in recipient mice. SCL-tTA/BCR-ABL transgenic mice were crossed with GFP transgenic mice to facilitate tracking of transplanted cells. Only LSK cells, but not CMP or GMP, from BM and spleen of BCR-ABL expressing mice were capable of generating CML-like disease and long term engraftment (>16 weeks) in recipient mice. The leukemic phenotype of donors was recapitulated in recipient mice; and leukemia could be transplanted to secondary and tertiary recipients. Further analysis revealed that the subpopulation of cells with a LT-HSC phenotype (Flt3-CD150+CD48-) within the LSK population was capable of generating CML-like disease and long term engraftment in recipient mice. Consistent with the diminished LT-HSC numbers demonstrated by flow cytometry, the frequency of functional HSC within the LSK population, as measured by competitive repopulation limiting dilution assays, was reduced in BCR-ABL expressing mice (1 in 234) compared to control mice (1 in 14). Interestingly, not all transplanted mice with long-term engrafted BCR-ABL-expressing cells developed leukemia. We determined that 1 in 6 Flt3-CD150+CD48- LSK cells possessed repopulation activity, whereas only 1 in 80 cells was capable of initiating leukemia in transplanted mice within 20 weeks, indicating that only a subset of BCR-ABL+ cells with long-term repopulating potential has leukemia-initiating capacity. In summary, BCR-ABL expression is associated with significant reduction in LT-HSC and expansion of MPP and GMP in the BM, and a marked increase in LT-HSC, MPP, CMP and GMP in the spleen of transgenic mice. Reduced LT-HSC numbers in BM may be explained by increased proliferation of BCR-ABL-expressing HSC and their enhanced egress from the BM to extramedullary locations such as the spleen. BCR-ABL expressing LT-HSC demonstrated long term engraftment and secondary transplantation capacity. However, only a fraction of BCR-ABL-expressing long-term repopulating cells has leukemia-initiating capacity, suggesting that additional cell intrinsic or extrinsic factors besides BCR-ABL expression may play a role in determining their leukemogenic potential. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1212-1212
Author(s):  
Bin Zhang ◽  
Yin Wei Ho ◽  
Qin Huang ◽  
Claudia Huettner ◽  
Ravi Bhatia

Abstract Abstract 1212 Specialized microenvironmental niches are essential for hematopoietic stem cell (HSC) lodgment and maintenance. However the niche interactions of leukemia stem cells (LSC) are largely unknown. Targeted expression of the BCR-ABL gene in murine hematopoietic stem and progenitor cells (HSPC), via a Tet-regulated SCL promoter, results in development of a chronic phase CML-like disorder (Blood 105:.324, 2005). We have employed this SCL-tTA-BCR/ABL mouse model to investigate the characteristics of LSC in CML. BCR-ABL transgenic mice were crossed with GFP transgenic mice to facilitate tracking of transplanted cells. We have reported that LSC capacity is restricted to a population of cells with LT-HSC phenotype (LSK Flt3-CD150+CD48-). BCR-ABL expression is associated with reduced numbers of LT-HSC in the BM and greatly increased numbers of LT-HSC in the spleen compared with controls(Blood 2009, 114: 858). These observations suggest that CML LT-HSC demonstrate altered niche requirements compared to normal LT-HSC. We therefore conducted additional studies to investigate whether abnormal localization of CML LT-HSC was related to reduced homing and/or reduced retention in the BM microenvironment. To evaluate LT-HSC homing, BCR-ABL+ and control LT-HSC (10,000 cells/mouse) were labeled with CFSE and injected by tail vein injection into wild type mice irradiated at 900cGy and CFSE+ cells in the BM and spleen of recipient mice were evaluated 4h after injection. We observed 32% reduction in homing of BCR-ABL+ LT-HSC to the BM of recipient mice compared to control LT-HSC (p=0.04), with similar homing to the spleen. To study trafficking from BM to extramedullary sites, BCR-ABL+ and control LT-HSC were injected directly into the right femur of irradiated congenic mice (1000 cells/mouse). Recipient mice were euthanized 2 and 4 weeks after injection and localization of LT-HSC, progenitors and WBC in the right femur, the contralateral (left) femur, and spleen analyzed by flow cytometry. We observed 4.9-fold increased numbers of BCR-ABL+ LT-HSC compared with control LT-HSC in the spleen(p=0.008) and 60% decreased numbers of BCR-ABL+ LT-HSC in the marrow compared with control LT-HSC at 4 weeks post-injection(p=0.048). Increased egress from BM to spleen was not related to BM hypercellularity. These results are consistent with enhanced egress of BCR-ABL+ LT-HSC from the BM to the spleen and/or enhanced growth in the spleen. No significant differences in expression of α4, α5, and α6 integrin and CD44 expression were seen in LT-HSC from the spleen and BM of BCR-ABL+ and control mice. A small population of β7 integrin expressing LT-HSC was seen in BM from BCR-ABL+ mice. Adhesion of LT-HSC from the spleen and BM of BCR-ABL+ and control mice to fibronectin coated wells was evaluated. LT-HSC from BM of BCR-ABL+ mice showed reduced adhesion to fibronectin after 2 hours(43±3%) compared to LT-HSC from control mice (57±3%, p=0.004), indicating impaired α4β1 and α5β1 integrin receptor function despite normal levels of receptor expression. BCR/ABL+ LT-HSC cells demonstrated higher CXCR4 expression and enhanced migration to CXCL12 (SDF-1) in a 3-hour transwell migration assay(20±5%), compared to control LT-HSC (9±3%, p=0.04). CXCL12-induced migration of BCR/ABL+ LT-HSC was completely blocked by the CXCR4 antagonist AMD3100. ELISA analysis of CXCL12 levels revealed 2.5-fold reduction in BM supernatants and 1.4-fold increase in splenic supernatants from BCR/ABL+ mice compared to control mice. We conclude that BCR-ABL expression results in significant reduction in LT-HSC homing to BM niches and markedly increased egress of LT-HSC from BM to the spleen through a combination of both intrinsic defects in LT-HSC adhesion and migration and leukemia-associated alterations in the BM and splenic microenvironments. Our results indicate LT-HSC-niche interactions are markedly perturbed in CML, potentially contributing to deregulated stem cell growth. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5165-5165
Author(s):  
Stephanie Thiant ◽  
Moutuaata M.Moutuou ◽  
Philippe Laflamme ◽  
Radia Sidi Boumedine ◽  
Fanny Larochelle ◽  
...  

Abstract PURPOSE: Chronic myelogenous leukemia (CML) is a disorder affecting early hematopoietic stem cells (HSC) and is characterized by excessive proliferation and accumulation of myeloid progenitors and progeny in the periphery. During the chronic phase of the disease, CML patients are normally at low risk of developing infections but such complications tend to rise during the progression of the disease. Gleevec (imatinib mesylate) is currently administered as first line therapy for patients with Philadelphia chromosome-positive CML. Despite the relative high specificity of tyrosine kinase inhibitor (TKI) treatment towards the BCR-ABL fusion protein, off-target multikinase inhibitory effects occur and can interfere with normal hematopoiesis. This study was conducted to evaluate how myeloid and lymphoid immune homeostasis are affected by Imatinib mesylate. METHODS: Healthy volunteer donors (n=25) and CML patients were recruited during their first visit at our CML clinic. Seven CML patients were treated with Imatinib (400mg). The median time of Gleevec treatment was 2.9 years (range: 0.5-10.9). The median time of remission post TKI was 1.1 years (range: 0.3-3). Phenotypic analysis of dendritic cell (DCs) subsets: plasmacytoid (pDCs) and myeloid type 1, 2 and 3 (mDC1, mDC2, mDC3) were evaluated by flow cytometry. Percentage and absolute numbers of naive and memory CD4+ and CD8+ T cells, NK cells and B cells were also evaluated. DCs were differentiated from purified CD34+ cells culturedwith GM-CSF (800 U/ml) or Flt3-L (50ng/ml), IL-4 (10 U/ml) and TNFa (50 U/ml), in the presence of varying concentrations of Imatinib mesylate (0 to 5µM/mL). TCR and IL-7 signaling were evaluated based on ERK-phosphorylation (-p) and STAT5-p after incubation with 3µM of Imatinib. RESULTS AND CONCLUSION: At diagnosis, several CML patients have a deficit in DCs resulting from a severe skewing affecting BM progenitor cells. After initiating Gleevec therapy, normalization of stem cell progenitors occurs but DC counts remain well below normal levels in all CML patients. We demonstrated a direct and dose dependent interference of Imatinib on GM-CSF and Flt3-L pathways for DC differentiation from CD34+ stem cells. For T lymphocytes, Imatinib interfered with TCR and IL-7 signaling through the inhibition of ERK and STAT5 phosphorylation respectively. The failure to maintain adequate numbers of DCs combined to diminished homeostatic response to cytokines and TCR stimuli explains T cell lymphopenia in these patients. Such immune dysfunction is at least in part responsible for infectious complications that are often increased in patients treated with Imatinib. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3149-3149 ◽  
Author(s):  
Yue Wei ◽  
Hong Zheng ◽  
Yu Jia ◽  
Naran Bao ◽  
Shan Jiang ◽  
...  

Abstract Myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML)aremyeloid neoplasms characterized by abnormal bone marrow hematopoiesis and increased risk of transformation to acute myelogenous leukemia (AML). Epigenetic dysregulation and inflammatory hyper-activation have been recognized as key molecular abnormalities in the bone marrow (BM) hematopoietic stem and progenitor cells (HSPC) of MDS and CMML, which implies that key modulators of epigenetic and inflammatory regulation play an important role in the pathophysiology of these diseases, which could also serve as effective therapeutic targets. We recently identified such a candidate molecule: the histone demethylase KDM6B (JMJD3). We demonstrated that KDM6B is significantly overexpressed in the BM HSPCs of patients with MDS and CMML, and the overexpression of KDM6B mediates aberrant epigenetic activation of innate immune/inflammatory signals and consequent differentiation skewing in BM HSPCs of MDS, which can be reversed by targeting KDM6B. Importantly, systematic analysis of the global transcriptomic and genomic data of patients indicates that, although KDM6B is overexpressed in MDS and CMML, genetic lesions in this gene are very rare, and higher KDM6B expression correlates with TET2 mutation. These results imply that constitutive expression of KDM6B potentially interacts with more common genetic lesions during the development of MDS and CMML. To further investigate the effects of KDM6B overexpression on hematopoiesis and its role in myeloid disorders, we developed a novel hematopoietic KDM6B transgenic (Tg) mouse model that overexpresses KDM6B under the control of the murine hematopoietic specific Vav promoter (Vav-KDM6B). Long-term monitoring of the peripheral blood counts of the mice indicates that, although younger Vav-KDM6B mice display only minor changes in whole white blood cells (WBC), monocytes, and platelets, aged KDM6B mice (>1 year old) have significant increases of WBC (by 22%, p<0.05) and monocyte counts (by 34%, p<0.01). In BM biopsies, the megakaryocytic progenitors of Vav-KDM6B mice specifically possess dysplastic morphology. Analysis of BM HSPCs revealed a tendency of increased numbers of long-term hematopoietic stem cells (LT-HSC) in Vav-KDM6B mice. Because MDS and CMML are associated with a pro-inflammatory BM microenvironment, we applied low-dose lipopolysaccharide (LPS) treatment (6 µg/mouse) for 6 weeks to both Vav-KDM6B and control mice. After chronic immune stimulation, more significant decreases of peripheral red cell (RBC) count and hemoglobin were observed in LPS-treated Vav-KDM6B mice compared to others. In BM biopsies, we noted more dramatic increases of megakaryoblasts in LPS-treated Vav-KDM6B mice than treated wild-type mice. Increases of BM HSPCs were also detected in LPS-treated Vav-KDM6B mice, including LSK cells and LT-HSC populations. Furthermore, the LSK cells isolated from LPS-treated Vav-KDM6B mice consistently demonstrated increased serial plating capacity in methocult-supported colony formation assays. Taken together, these hematopoietic phenotypes observed in the LPS-treated Vav-KDM6B Tg mice indicate that KDM6B overexpression in combination with pro-inflammatory stimulation can accelerate the occurrence of the MDS- and CMML-like abnormalities in BM HSPCs. To gain more insight into the cellular and molecular impacts of KDM6B overexpression on BM HSPCs, detailed analysis, including in vivo repopulating capacity assays as well as gene expression profiling, are being performed. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 3 (3) ◽  
pp. 419-431 ◽  
Author(s):  
Fang Dong ◽  
Haitao Bai ◽  
Xiaofang Wang ◽  
Shanshan Zhang ◽  
Zhao Wang ◽  
...  

Abstract The cell of origin, defined as the normal cell in which the transformation event first occurs, is poorly identified in leukemia, despite its importance in understanding of leukemogenesis and improving leukemia therapy. Although hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) were used for leukemia models, whether their self-renewal and differentiation potentials influence the initiation and development of leukemia is largely unknown. In this study, the self-renewal and differentiation potentials in 2 distinct types of HSCs (HSC1 [CD150+CD41−CD34−Lineage−Sca-1+c-Kit+ cells] and HSC2 [CD150−CD41−CD34−Lineage−Sca-1+c-Kit+ cells]) and 3 distinct types of HPCs (HPC1 [CD150+CD41+CD34−Lineage−Sca-1+c-Kit+ cells], HPC2 [CD150+CD41+CD34+Lineage−Sca-1+c-Kit+ cells], and HPC3 [CD150−CD41−CD34+Lineage−Sca-1+c-Kit+ cells]) were isolated from adult mouse bone marrow, and examined by competitive repopulation assay. Then, cells from each population were retrovirally transduced to initiate MLL-AF9 acute myelogenous leukemia (AML) and the intracellular domain of NOTCH-1 T-cell acute lymphoblastic leukemia (T-ALL). AML and T-ALL similarly developed from all HSC and HPC populations, suggesting multiple cellular origins of leukemia. New leukemic stem cells (LSCs) were also identified in these AML and T-ALL models. Notably, switching between immunophenotypical immature and mature LSCs was observed, suggesting that heterogeneous LSCs play a role in the expansion and maintenance of leukemia. Based on this mouse model study, we propose that acute leukemia arises from multiple cells of origin independent of the self-renewal and differentiation potentials in hematopoietic stem and progenitor cells and is amplified by LSC switchover.


Sign in / Sign up

Export Citation Format

Share Document