scholarly journals Cell Competition and Microenvironment in the Development of Myeloproliferative Neoplasms

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2485-2485
Author(s):  
Melissa Castiglione ◽  
Huichun Zhan

Introduction The myeloproliferative neoplasms (MPNs) are clonal stem cell disorders characterized by hematopoietic stem/progenitor cell (HSPC) expansion. The acquired kinase mutation JAK2V617F plays a central role in MPNs. Both JAK2 wild-type (WT) clones and JAK2V617F-mutant clones co-exist in many MPN patients. Little is known about earliest steps of MPN disease initiation and how the mutant cells expand over WT cells. In this work, we tested the hypothesis that competition between WT and JAK2V617F-mutant cells acts to prevent mutant cell expansion in MPNs, but that a JAK2V617F-bearing microenvironment can overcome this competition, leading to the development of a MPN. Methods JAK2V617F Flip-Flop (FF1) mice and Tie2-Cre mice were crossed to generate a strain in which human JAK2V617F is expressed specifically in hematopoietic cells and vascular endothelial cells (Tie2FF1). Results First, we isolated JAK2WT and JAK2V617F-mutant CD45+CD201+CD150+CD48- (here termed HSPCs) from either WT mice or Tie2FF1 mice and performed a series of in vitroco-culture assays. We found that JAK2V617F HSPCs displayed a higher proliferation rate than JAK2WT HSPCs when cultured separately. In contrast,when placed in direct co-culture, proliferation of JAK2WT HSPCs was greatly increased that no significant difference between WT and mutant HSPCs was detected. Similar results were also obtained when using a transwell co-culture system, suggesting that cell-cell contact was not required for the competitive co-existence we observed in mixed JAK2WT / JAK2V617F HSPC cultures in vitro. (Figure 1) Next, we performed marrow transplantation assays to assess the competition between JAK2WT and JAK2V617F HSPCs in vivo. We found that, when 50% JAK2V617F marrow cells and 50% JAK2WT marrow cells were transplanted together into lethally irradiated WT mice, the JAK2V617F donor cells had no engraftment advantage over JAK2WT donor cells and the recipient mice had normal blood cell counts during more than 4-month follow up. This result contrasts our and other reports that a JAK2V617F-positive MPN develops following transplantation of 100% JAK2V617F-mutant donor cells into WT recipients. These observations suggest that the presence of WT cells can alterthe behavior of JAK2V617F-mutant stem cells in vivo. This conclusion was further supported by in vivoBrdU labeling experiments, which demonstrated that the proliferation rate of JAK2V617F-mutant Lin-cKit+Sca1+CD150+CD48-(here termed HSCs)was 2-fold higher than JAK2WT HSCs when transplanted separatelyinto lethally irradiated WT recipients; however, when transplanted together, WT HSCs displayed a significantly higher proliferation rate (3-fold) than mutant HSCs. (Figure 2) Endothelial cells (ECs) are an essential component of the hematopoietic niche and many studies have established that a significant number of ECs in MPN patients carry the JAK2V617F mutation. Using the same Tie2FF1 murine model, we previously reported that a JAK2V617F-bearing vascular niche promotes the expansion of JAK2V617F HSPCs over JAK2WT HSPCs, and the level of Chemokine (C-X-C motif) ligand 12 (CXCL12) was upregulated in JAK2V617F-mutant ECs compared to WT ECs. Cell cycle analysis using Hoechst33342 and Pyronin Y staining demonstrated that, while there were no differences in WT and mutant HSPC cell cycle status when transplanted into a WT vascular niche, JAK2V617F-mutant HSPCs were less quiescent (more cycling) than WT HSPCs when transplanted into a JAK2V617F-mutant vascular niche. (Figure 3) To test the hypothesis that JAK2V617F-mutant vascular niche overcomes the competition between WT and mutant cells to promote mutant HSPC expansion through increased CXCL12 level, we used the same competitive repopulation assay as in Figure 2 where both JAK2WT and JAK2V617F marrow cells were injected together into lethally irradiated WT recipient. At 12wks post transplantation, the recipient mice were treated with a single dose of CXCL12 (400ng, intravenously) before BrdU labeling. We found that CXCL12 treatment stimulated JAK2V617F-mutant HSC proliferation to a greater extent than JAK2WT HSPCs. (Figure 4) Conclusions Cell competition between WT and JAK2V617F-mutant cells prevents mutant stem cells from expansion, but a mutant microenvironment (e.g. JAK2V617F-mutant vascular niche) can overcome this competition to promote mutant stem cell expansion and the development of a murine MPN. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 952-952 ◽  
Author(s):  
Huichun Zhan ◽  
Chi Hua Sarah Lin ◽  
Yarden Segal ◽  
Kenneth Kaushansky

Abstract Introduction The myeloproliferative neoplasms (MPNs) are clonal stem cell disorders characterized by hematopoietic stem/progenitor cell (HSPC) expansion and overproduction of mature blood cells. The acquired mutation JAK2V617F plays a central role in these disorders. The mechanisms responsible for MPN HSPC expansion are not fully understood, limiting the effectiveness of current treatments. Endothelial cells (ECs) carrying the JAK2V617F mutation can be detected in patients with MPNs. In this work we test the hypothesis that the JAK2V617F-mutant vascular niche promotes JAK2V617F clonal expansion in MPNs. Methods JAK2V617F Flip-Flop (FF1) mice (Radek Skoda, Switzerland) and Tie2-Cre mice (Mark Ginsberg, UC San Diego) were crossed to generate a strain in which human JAK2V617F is expressed specifically in hematopoietic cells and ECs (Tie2/FF1). Animal experiments were performed in accordance with the Institutional Animal Care and Use Committee guideline. Results Primary lung ECs (CD45-CD31+) were isolated from Tie2/FF1 mice (JAK2V617F ECs) and age-matched littermate control mice (JAK2WT EC). JAK2V617F ECs proliferated to a greater extent than JAK2WT ECs. A vascular tube formation assay was performed as a measure of in vitro angiogenesis and revealed that JAK2V617F ECs had significantly increased angiogenesis compared to JAK2WT EC. (Figure 1) To examine the role of JAK2WT and JAK2V617F ECs in MPN hematopoiesis in vitro, marrow Lin-cKit+ HSPCs were isolated from Tie2/FF1 and age-matched littermate controls and cultured on a feeder layer of JAK2WT or JAK2V617F ECs under serum-free conditions. While there was no difference between JAK2V617F and JAK2WT HSPC proliferation when co-cultured with JAK2WT EC, the JAK2V617F HSPC displayed a relative growth advantage over the JAK2WT HSPC (1.24-fold, p = 0.041) when co-cultured on JAK2V617F EC. Next, the effects of JAK2V617F-bearing vascular niche on MPN hematopoiesis were studied in vivo using marrow transplantation assays. First, wild-type CD45.1 marrow cells were transplanted into lethally irradiated (950cGy) 8-14 week old Tie2/FF1 mice or controls (CD45.2) (n=7 in each group). During a >8-month follow up, all wild-type recipients displayed full donor engraftment, while 4 of 7 Tie2/FF1 recipient mice displayed recovery of JAK2V617F-mutant hematopoiesis (mixed donor/recipient chimerism), suggesting that the JAK2V617F-mutant vascular niche may protect the JAK2V617F-mutant HSPCs from the lethal irradiation. Although marrow and splenic hematopoietic progenitors were significantly increased in the Tie2/FF1 recipients, there was no difference in peripheral blood (PB) cell count or PB hematopoietic progenitor cell numbers between the Tie2/FF1 mice and control mice. (Figure 2) Second, we performed a competitive marrow transplantation experiment in which 5 x 105 CD45.2 donor marrow cells from Tie2/FF1 mice were injected intravenously together with 5 x 105 competitor CD45.1 wild-type marrow cells intro lethally irradiated Tie2/FF1 mice or control mice (CD45.2) (n=5 in each group). During a 4-month follow up, Tie2/FF1 recipients displayed lower PB CD45.1 (i.e. wild-type donor) chimerism than the control mice (p = 0.025), suggesting that the mutant vascular niche in Tie2/FF1 promoted the JAK2V617F clonal maintenance/expansion and/or inhibited the wild-type hematopoiesis. By 18 weeks post-transplant, Tie2/FF1 recipients developed a MPN phenotype with elevated WBC count (22.7 vs. 9.85 x 109/L, p = 0.049), platelet count (5336 vs. 1029 x 109/L, p = 0.033), increased splenomegaly, increased total marrow and splenic hematopoietic progenitors, and increased CD150+CD48- E-SLAM cells (a highly enriched HSPC population) in both marrow (5-fold, p = 0.0004) and spleen (5.5-fold, p = 0.000004) compared to the control mice. To begin to understand the EC signals responsible for JAK2V617F clonal expansion and MPN pathogenesis, qRT-PCR of purified marrow ECs identified up-regulation of MPL and CXCL12 in both arterial (CD45-CD31+Sca1+) and sinusoidal (CD45-CD31+Sca1-) marrow ECs in Tie2/FF1 mice compared to controls. Conclusions Our study establishes that JAK2V617F-bearing ECs form an important part of the MPN HSPC niche and contribute to mutant stem/progenitor cell expansion both in vitro and in vivo. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1599-1599 ◽  
Author(s):  
Niccolò Bartalucci ◽  
Costanza Bogani ◽  
Serena Martinelli ◽  
Carmela Mannarelli ◽  
Jean-Luc Villeval ◽  
...  

Abstract Background and Aims A gain-of-function mutation in Janus kinase 2 (JAK2V617F) is at the basis of the majority of chronic myeloproliferative neoplasms (MPN). The dual JAK1/JAK2 inhibitor ruxolitinib (ruxo) determined rapid and sustained responses in splenomegaly and symptomatic improvement in patients with myelofibrosis (MF), supporting the central role of dysregulated JAK2 signaling. Enhanced activation of other downstream pathways including the PI3K/mTOR pathway has been documented as well. We previously reported (Bogani et al, PlosOne 2013;8:54828) that targeting mTOR by the allosteric inhibitor RAD001 resulted in inhibition of JAK2VF mutated cells and produced clinical benefits in a phase I/II trial (Guglielmelli et al, Blood 2011;118:2069). In this study we evaluated the effects of BKM120, a specific PI3K inhibitor, alone and in combination with ruxolitinib, in in-vitro and in-vivo MPN models. Methods To evaluate cell proliferation, colony formation, apoptosis, cell cycle and protein phosphorylation status we used mouse BaF3 and BaF3-EPOR cells expressing wild type (WT) or VF mutated JAK2, the human VF-mutated HEL and SET2 cell lines, and primary MPN CD34+ cells from patients with MF or polycythemia vera (PV). Effect of drug combination was analyzed according to Chou and Talalay calculating the combination index (CI); a CI <1 indicates synergistic activity. For in vivo studies we used two mouse models: (1) SCID mice receiving iv BaF3-EPOR VF-luciferase (luc) cells (gift of T. Radimerski) were randomized on day 6 to different treatment groups based on baseline luminescence. (2) C57Bl6/J JAK2 VF Knock-in mice were generated by insertion of the reversed JAK2V617F exon 13 sequence; mating with Vav-Cre transgenic mice activates the VF allele producing a MPN phenotype in progenies with VF heterozygous expression (Hasan et al, Blood 2013;Epub). Mice were treated for 15 days, then blood, spleen and bone marrow cells were analyzed. Results We found that BKM120 preferential inhibited BAF3 VF and BaF3-EpoR VF cells (IC50: 364±200nM and 1100±207nM, respectively) compared to their respective WT counterpart (5300±800nM and 3122±1000nM: p<.05). HEL and SET2 cells resulted also sensitive to BKM120 (2000±500nM and 1000±300nM). Interestingly we found that BKM120 significantly increased G2/M phase and decreased S phase of cell cycle (p<.01) and induced apoptosis (IC50, SET2=10µM, BaF3-EPOR VF=1.8 µM). Western blot analysis showed marked reduction of phospho-mTOR and its target phospho-4EBP1 as well as downregulation of phospho-STAT5 at 6 and 24h of treatment. BKM120 impaired colony formation from MF and PV CD34+ cells at doses 2 to 8-fold lower than healthy controls (p<.01). BKM120 strongly inhibited EEC colony growth from PV pts (IC50, 9±4nM). Co-treatment of BKM120+ruxo resulted in synergistic inhibition of proliferation of SET2 (median CI=0.45) and BaF3-EPOR VF (median CI=0.8) cells. Triple combinations including BKM120/ruxo plus either RAD001 (Torc1 inhibitor) or PP242 (Torc1/2 inhibitor) resulted highly synergistic (median CI=0.27 and 0.52) to indicate the importance of complete mTOR inhibition. BKM120 at 45mpk and 60mpk increased mean lifespan of BaF3 VF luc mouse model from 21d in control mice to 27.2d and 28d in BMK120 treated mice. In KI mice, co-treatment with 60mpk BKM120 + 60mpk ruxo resulted in improvement of splenomegaly (median spleen weight: 1.4, 0.82, 0.8 and 0.6 g respectively for controls, 60mpk BKM120, 60mpk ruxo and 60mpk BKM120+60mpk ruxo) and reduction of leukocytosis and reticulocyte count. The level of phosho-STAT5 and -4EBP1 in the spleen was significantly reduced in mice receiving BKM120+ruxo as compared to single drug treatment. We finally analyzed the effects of BKM120+/-ruxo on the in-vitro clonogenic growth of BM cells from VF and WT KI mice mixed in a 1:1 ratio. The proportion of VF-positive colonies resulted reduced in a dose dependent manner by 19%, 33% and 44% (p<.03) compared to controls with 50nM, 100nM and 300nM BKM120 respectively. A 25% and 39% of VF-positive colonies reduction was achieved with 50nM and 100nM ruxolitinib. The combined treatment with 100nM BKM120 + 50nM ruxo resulted in a 50% decrease of the number of mutated colonies (p<.02). Conclusions Inhibition of PI3K by BKM120 alone and combined with JAK2 inhibitor ruxolitinib resulted in enhanced activity in preclinical models of MPN, providing a rationale for the ongoing combination clinical trial. Disclosures: Vannucchi: Novartis: Membership on an entity’s Board of Directors or advisory committees.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 20-20
Author(s):  
James Bartram ◽  
Baobao (Annie) Song ◽  
Juying Xu ◽  
Nathan Salomonis ◽  
H. Leighton Grimes ◽  
...  

Abstract Hematopoietic stem cells are endowed with high regenerative potential but their actual self-renewal capacity is limited. Studies using the H2B-retention labeling system show HSC functional decline at each round of division (Qiu, Stem Cell Reports 2014). We have shown that mitochondria drive HSC functional decline with division history after transplantation (Cell Stem Cell 2020). Here we examined the link between mitochondrial metabolism, in vivo division at steady state, and HSC functions using the GFP label-Histone 2B (GFP-H2B) mouse model driven by a doxycycline-inducible promoter. Five months after doxycycline removal, mitochondrial membrane potential (MMP) was examined using TMRE in HSC with varying GFP intensity. HSC were separated into an H2B-labeled retention population and an H2B-labeled population. Interestingly, within the H2B-labeled retention population, HSC could be further subdivided into GFP high, medium, and low. MMP increased in a stepwise fashion with GFP dilution in HSC. We noted the presence of 2 TMRE peaks within each GFP high and medium populations leading to 5 populations: GFP-high;MMP-low (G1), GFP-high;MMP-high (G2), GFP-medium;MMP-low (G3), GFP-medium;MMP-high (G4), GFP-low;MMP-high (G5). We examined the repopulation activity of each population in a serial competitive transplant assay. G1 and G2 maintained higher peripheral blood chimerism up to 24 weeks post-transplant than G3 and G4. G5 did not engraft at all. However, only G1 reconstituted high frequency of HSC in primary recipients. In secondary recipients, G1, G2, G3 but not G4 gave rise to positive engraftment. Interestingly, G1 and G2 grafts showed myeloid/lymphoid balanced engraftment whereas the G3 graft was myeloid-bias, suggesting that myeloid skewing can be acquired upon HSC division. We further examined lineage fate maps of bone marrow cells derived from G1 or G3 population in vivo, using single cell RNA sequencing, 10X genomics. Surprisingly, G3-derived bone marrow cells displayed a distinct myeloid cell trajectory from G1-derived bone marrow cells, in which G3 gave rise to increased immature neutrophils but fewer myeloid precursors. Remarkably, each lineage population derived from G3 donor cells had different gene expression signatures than those derived from G1 donor cells. Therefore, HSC that have divided in vivo in the same bone marrow microenvironment are intrinsically and molecularly different such that not only do they exhibit lineage potential differences but they also produce progeny that are transcriptionally different. These findings imply that cellular division rewires HSC and that this rewiring is passed down to their fully differentiated progeny. When G1 and G3 single HSC were cultured in-vitro, G1 had a slower entry into cell-cycle which has been associated with increased stemness. Additionally, when single HSC from G1 and G3 were assessed for their multipotency in a lineage differentiation assay, G1 HSC had a higher propensity to produce all four myeloid lineages (megakaryocytes, neutrophils, macrophages, and erythroid), further supporting increased stemness in G1 compared to G3 HSC. Finally, HSC from G1, G2, G3 and G4 populations carried mitochondria that were morphologically different, and express distinct levels of Sca-1, CD34 and EPCR, with Sca-1 high, CD34-, EPCR+ cells more enriched in G1. In summary, this study suggests that HSC transition into distinct metabolic and functional states with division history that may contribute to HSC diversity and functional heterogeneity. It also suggests the existence of a cell-autonomous mechanism that confers HSC divisional memory to actively drive HSC functional heterogeneity and aging. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1468-1468
Author(s):  
Haotian Zhang ◽  
Melissa Castiglione ◽  
Lei Zheng ◽  
Huichun Zhan

Abstract Introduction Disease relapse after allogeneic stem cell transplantation is a major cause of treatment-related morbidity and mortality in patients with myeloproliferative neoplasms (MPNs). The cellular and molecular mechanisms for MPN relapse are not well understood. In this study, we investigated the role of cell competition between wild-type and JAK2V617F mutant cells in MPN disease relapse after stem cell transplantation. Methods JAK2V617F Flip-Flop (FF1) mice (which carry a Cre-inducible human JAK2V617F gene driven by the human JAK2 promoter) were crossed with Tie2-cre mice to express JAK2V617F specifically in all hematopoietic cells and vascular endothelial cells (Tie2FF1), so as to model the human diseases in which both the hematopoietic stem cells and endothelial cells harbor the mutation. Results To investigate the underlying mechanisms for MPN disease relapse, we transplanted wild-type CD45.1 marrow directly into lethally irradiated Tie2FF1 mice or age-matched control mice(CD45.2). During a 6-7mo follow up, while all wild-type control recipients displayed full donor engraftment, ~60% Tie2FF1 recipient mice displayed recovery of the JAK2V617Fmutant hematopoiesis (mixed donor/recipient chimerism) 10 weeks after transplantation and developed a MPN phenotype with neutrophilia and thrombocytosis, results consistent with our previous report. Using CD45.1 as a marker for wild-type donor and CD45.2 for JAK2V617F mutant recipient cells, we found that the wild-type HSCs (Lin -cKit +Sca1 +CD150 +CD48 -) were severely suppressed and the JAK2V617F mutant HSCs were significantly expanded in the relapsed mice; in contrast, there was no significant difference between the wild-type and mutant HSC numbers in the remission mice. (Figure 1) Cell competition is an evolutionarily conserved mechanism in which "fitter" cells out-compete their "less-fit" neighbors. We hypothesize that competition between the wild-type donor cells and JAK2V617F mutant recipient cells dictates the outcome of disease relapse versus remission after stem cell transplantation. To support this hypothesis, we found that there was no significant difference in cell proliferation, apoptosis, or senescence between wild-type and JAK2V617F mutant HSPCs in recipient mice who achieved disease remission; in contrast, in recipient mice who relapsed after the transplantation, wild-type HSPC functions were significantly impaired (i.e., decreased proliferation, increased apoptosis, and increased senescence), which could alter the competition between co-existing wild-type and mutant cells and lead to the outgrowth of the JAK2V617F mutant HSPCs and disease relapse. (Figure 2) To understand how wild-type cells prevent the expansion of JAK2V617F mutant HSPCs, we established a murine model of wild-type and JAK2V617F mutant cell competition. In this model, when 100% JAK2V617F mutant marrow cells (from the Tie2FF1 mice) are transplanted alone into lethally irradiated wild-type recipients, the recipient mice develop a MPN phenotype ~4wks after transplantation; in contrast, when a 50-50 mix of mutant and wild-type marrow cells are transplanted together into the wild-type recipient mice, the JAK2V617F mutant donor cells engraft to a similar level as the wild-type donor cells and the recipient mice displayed normal blood counts during more than 4-months of follow up. In this model, compared to wild-type HSPCs, JAK2V617F mutant HSPCs generated significantly more T cells and less B cells in the spleen, and more myeloid-derived suppressor cells (MDSCs) in the marrow; in contrast, there was no difference in T, B, or MDSC numbers between recipients of wild-type HSPCs and recipients of mixed wild-type and JAK2V617F mutant HSPCs. We also found that program death ligand 1 (PD-L1) expression was significantly upregulated on JAK2V617F mutant HSPCs compared to wild-type cells, while PD-L1 expression on mutant HSPCs was significantly decreased when there was co-existing wild-type cell competition. These results indicate that competition between wild-type and JAK2V617F mutant cells can modulate the immune cell composition and PD-L1 expression induced by the JAK2V617F oncogene. (Figure 3) Conclusion Our study provides the important observations and mechanistic insights that cell competition between wild-type donor cells and JAK2V617F mutant recipient cells can prevent MPN disease relapse after stem cell transplantation. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 260-260 ◽  
Author(s):  
Jeffrey S. Miller ◽  
Yvette Soignier ◽  
Angela Panosskaltsis-Mortari ◽  
David McKenna ◽  
Chap Le ◽  
...  

Abstract We previously demonstrated that although autologous NK cell therapy after hematopoietic cell transplantation (HCT) is safe, it does not provide an anti-tumor effect. We hypothesize that the lack of NK cell inhibitory receptor (KIR) mismatching with autologous tumor cells limits anti-tumor efficacy. Allogeneic NK cell infusions may overcome this restriction. Here we tested haploidentical, related donor NK cell infusions in a non-transplant setting to establish safety with a goal of in vivo NK cell expansion. A low intensity outpatient immune suppressive regimen of fludarabine (Flu) alone was tested in 8 renal cell carcinoma patients. A higher intensity inpatient regimen of high-dose cyclophosphamide and Flu (Hi-Cy/Flu) was tested in 12 patients with poor prognosis AML, all with active leukemia and most who failed 1 or more standard theries. NK cells were enriched from donor apheresis products using CD3 depletion by the CliniMacs system. All patients received subcutaneous IL-2 after infusions. Patients who received the lower intensity Flu regimen showed transient persistence of donor cells, but no in vivo expansion (&lt;0.1%) using a PCR based chimerism assay of unique MHC class I alleles in the donor but not present in the recipient. There were no objective responses in renal cell carcinoma patients. In marked contrast, 6 of 7 AML patients who were evaluable for engraftment receiving Hi-Cy/Flu showed engraftment of donor cells, some for &gt; 28 days. Three of 6 patients had peak engraftment levels of 10%, 100% and 100% of PBMC in the first 2 weeks after the haploidentical infusion. The remaining three patients had lower levels of approximately 1%. Five of 12 patients have achieved a remission with this therapy. Compared to patients who failed to clear their leukemia, the remission patients had significantly higher proportions of circulating NK cells (35±8% vs. 1.5±0.4%, p=.001) and cytotoxicity against K562 targets (50±10% vs. 10±5% at E:T 20:1, p=0.01) suggesting that in vivo expansion was required to achieve efficacy. We next tested whether endogenous cytokines play a role in the in vivo expansion. Based on its established role in NK cell differentiation and on recent mouse studies, IL-15 was a likely candidate to drive homeostatic NK cell expansion. Flu patients showed only a slight increase in IL-15 after several days compared to pre-treatment levels. In marked contrast, AML patients had significant increases in plasma IL-15 after Hi-Cy/Flu therapy, which appeared before haploidentical cell infusions and was sustained for several weeks. There was an inverse correlation between the absolute lymphocyte count and the IL-15 concentration (r = −.62, p-value &lt; .0001). We conclude that Hi-Cy/Flu therapy allows for expansion of allogeneic NK cells in vivo in part through increased endogenous IL-15 concentrations, which can induce remissions in poor prognosis AML patients. For the first time, these findings suggest that haploidentical NK cells can persist and expand in vivo and may have a role in the treatment of AML used alone or as an adjunct to HCT. Future studies will address NK cell donor-recipient mismatch at KIR ligands, patient selection and other variables that impact on response.


2008 ◽  
Vol 49 (4) ◽  
pp. 437-443 ◽  
Author(s):  
Kanokporn Noy RITHIDECH ◽  
Marc GOLIGHTLY ◽  
Elbert WHORTON

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2617-2617
Author(s):  
Charu Gupta ◽  
Michelle Maria Araujo Cruz ◽  
Nidhi Jyotsana ◽  
Amit Sharma ◽  
Ramya Goparaju ◽  
...  

Abstract #Michael Heuser and Anuhar Chaturvedi share senior authorship Background: Isocitrate dehydrogenase-1 (IDH1) is mutated in about 6% of AML patients. Mutant IDH produces R-2-hydroxyglutarate (R-2HG), which induces histone and DNA hypermethylation through inhibition of epigenetic regulators, thereby linking metabolism to tumorigenesis. We recently reported that at comparable intracellular R-2HG levels, mice receiving transplants of IDH1 mutant cells died significantly earlier than R-2HG treated mice in the context of HOXA9 overexpression. This suggests oncogenic functions of mutant IDH1 beyond R-2HG production. We employed a splice variant of mutated IDH1 that does not produce R-2HG (IDH1mutantΔ7) to decipher R-2HG independent signaling pathways that may contribute towards leukemogenesis. Methods: Bone marrow cells from mice were immortalized with HoxA9, and IDH1wildtype (IDH1wt), IDH1mutant (IDH1mut), IDH1wildtypeΔ7 (IDH1wtΔ7) and IDH1mutΔ7, were constitutively expressed and the leukemogenic potential was evaluated in vivo. Intracellular R-2HG was measured by enantiomer-specific quantification. Deletion of exon 7 from IDH1mut leads to a frameshift that creates a premature stop codon in the 9th exon, finally producing a 119 amino acids truncated protein, IDH1mutΔ7. This splice variant does not produce increased levels of R-2HG. The signaling pathways were explored by immunoblotting and immunofluorescence. Results: Mice receiving cells with IDH1mutΔ7 had the same short latency to leukemia as mice receiving cells with full-length mutant IDH1, while IDH1wt and IDH1wtΔ7 cells died with significantly longer latency. The WBC count increased over time in IDH1mutΔ7 mice similar to IDH1mut mice, whereas WBC counts in IDH1wtΔ7 mice remained normal. IDH1mutΔ7 mice died from monocytic leukemia that was phenotypically and morphologically indistinguishable from IDH1mut mice. HoxA9 IDH1mutΔ7 cells were readily transplantable into secondary recipients. During in vivo cell cycle analysis, we observed that the proportion of cells in S/G2/M phases was significantly higher in bone marrow cells transduced with IDH1mut or IDH1mutΔ7 when compared to cells transduced with IDH1wt or CTL. These data suggest that mutant IDH1 enhances myeloproliferation even in the absence of R-2HG. To identify R-2HG independent signaling pathways mediated by the mutant IDH1 protein, we first analyzed the gene expression of important regulators of cell cycle, differentiation, cell signaling and transcription by quantitative RT-PCR. Several genes (Ccnd1, Slc2a, Hdac3, Tgif2,and c-myc) were upregulated in IDH1mut and IDH1mutΔ7 cells compared to IDH1wt cells. Interestingly, we found a specific up-regulation of Ctnnb1 and Nfkb genes in IDH1mutΔ7 cells over both IDH1mut and IDH1wt cells. We next validated our mRNA expression results by immunoblotting and found that NFKB and ERK signaling were upregulated in both IDH1mut and IDH1mutΔ7 compared to IDH1wt and IDH1wtΔ7 cells. Interestingly, the protein level of β-catenin, STAT3 and STAT5 were many fold higher in IDH1mutΔ7 compared to IDH1mut and IDH1wt cells. β-catenin is known to be transactivated via c-Src, which is phosphorylated by EGFR to promote β-catenin nuclear localization and signaling. We traced this pathway for its relevance in our cells and found that IDH1mutΔ7 cells indeed showed higher levels of both EGFR and c-Src phosphorylation compared to IDH1mut cells. We performed immunofluorescence and cellular fractionation for β-catenin and found it to be partially localized in the nucleus in IDH1mutΔ7 but not in IDH1mut cells. We also observed an up-regulated STAT3 phosphorylation in IDH1mutΔ7 cells over IDH1mut. Conclusions: In summary, mutant IDH1 activates ERK and NFKB signaling, which is attributed to both R-2HG dependent and independent mechanisms of leukemogenesis. Interestingly, IDH1mutΔ7 employs R-2HG independent EGFR/β-catenin and JAK/STAT signaling for oncogenesis. This R-2HG-independent leukemogenesis reveals a novel signaling dynamic of IDH1mut which should be evaluated for its therapeutic potential. Disclosures Ganser: Novartis: Membership on an entity's Board of Directors or advisory committees. Heuser:Astellas: Research Funding; Karyopharm: Research Funding; Novartis: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria, Research Funding; Janssen: Consultancy; StemLine Therapeutics: Consultancy; Bayer Pharma AG: Consultancy, Research Funding; Sunesis: Research Funding; BergenBio: Research Funding; Tetralogic: Research Funding; Daiichi Sankyo: Research Funding.


eLife ◽  
2017 ◽  
Vol 6 ◽  
Author(s):  
Sonal Nagarkar-Jaiswal ◽  
Sathiya N Manivannan ◽  
Zhongyuan Zuo ◽  
Hugo J Bellen

Here, we describe a novel method based on intronic MiMIC insertions described in Nagarkar-Jaiswal et al. (2015) to perform conditional gene inactivation in Drosophila. Mosaic analysis in Drosophila cannot be easily performed in post-mitotic cells. We therefore, therefore, developed Flip-Flop, a flippase-dependent in vivo cassette-inversion method that marks wild-type cells with the endogenous EGFP-tagged protein, whereas mutant cells are marked with mCherry upon inversion. We document the ease and usefulness of this strategy in differential tagging of wild-type and mutant cells in mosaics. We use this approach to phenotypically characterize the loss of SNF4Aγ, encoding the γ subunit of the AMP Kinase complex. The Flip-Flop method is efficient and reliable, and permits conditional gene inactivation based on both spatial and temporal cues, in a cell cycle-, and developmental stage-independent fashion, creating a platform for systematic screens of gene function in developing and adult flies with unprecedented detail.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 485-485
Author(s):  
Huichun Zhan ◽  
Yupo Ma ◽  
Kenneth Kaushansky

Abstract Introduction The myeloproliferative neoplasms (MPNs), including essential thrombocythemia (ET), are characterized by unregulated hematopoietic stem cell (HSC) expansion and overproduction of blood cells. The JAK2V617F mutation can be found in majority of MPN patients, but the mechanisms responsible for the MPN stem cell expansion remain incomplete. One hallmark feature of MPNs is megakaryocyte (MK) hyperplasia. We hypothesize that JAK2V617F -mutant MKs contribute to MPN pathogenesis by promoting HSC expansion. Methods JAK2V617F Flip-Flop (FF1) mice and Pf4-Cre mice (from Radek Skoda, Switzerland) were crossed to generate a MK cell lineage-specific human JAK2V617F knock-in mouse strain (Pf4/FF1). Animal experiments were performed in accordance with the Institutional Animal Care and Use Committee guideline. Mice were sacrificed at 28 wk of age. Bone marrow and spleen cell flow cytometry analysis was performed on a FACSAriaTM III (BD). Mouse methylcellulose media and Megacult-C media (Stem Cell Technologies) were used to assay colony formation. The TaqMan¨ Gene Expression Assay (Applied Biosystems) was used for quantitative real-time polymerase chain reaction (qRT-PCR) to verify differential expression of fibroblast growth factor 1 (FGF1), platelet factor 4 (Pf4), transforming growth factor beta 1 (TGFb-1), chemokine (C-X-C motif) ligand 12 (CXCL12). For HSC transplantation experiments, wild-type CD45.1+ recipient mice (B6.SJL, Taconic) were lethally irradiated (950 cGy) and then received 5 x 106 nucleated marrow cells from Pf4/FF1 or age-matched littermate control mice (CD45.2). Results Thrombocytosis was observed in Pf4/FF1 mice at 16wk of age. At 28wk there was a significant increase in platelet count (1406 vs. 808 x 109/L, p = 0.0009) in Pf4/FF1 mice but no change was noted in hemoglobin or white blood cell count compared with age-matched littermate controls. Splenomegaly was observed in 28wk old Pf4/FF1 mice compared to controls (spleen weight 0.101 vs. 0.070 gram, p = 0.040). Marrow cell colony assays demonstrated an increase in total hematopoietic progenitors (1.8-fold, p = 0.007) and in MK-committed progenitors (1.9-fold, p = 0.00009) in Pf4/FF1 mice compared to controls (Figure 1). Marrow cell flow cytometry analysis revealed that CD45+ EPCR+ CD48- CD150+ (E-SLAM) cells, a highly purified HSC population, were increased 3.2-fold (p = 0.028, n=4) in Pf4/FF1 mice compared to control mice, although the cells did not express JAK2V617F as determined by RT-PCR for FF1. CD41+ MK cells were also increased in Pf4/FF1 mice (Figure 2). Histologic analysis of marrow sections revealed an increased megakaryopoiesis with more MKs adjacent to the endosteal surface in Pf4/FF1 mice than in controls. In addition, there were dilated marrow sinusoids in Pf4/FF1 mice and sinusoidal endothelial cells (EC) were surrounded by MKs that conformed to the EC morphology (Figure 3). No marrow or spleen fibrosis was observed at 28 wk of age. To more directly test the effects of JAK2V617F -mutant MKs on HSCs, marrow cells from 28-wk old Pf4/FF1 mice or controls (CD45.2) were transplanted into lethally irradiated congenic CD45.1 recipients (n = 5 in each group). By 12 wks post transplantation, recipients of Pf4/FF1 marrow had already developed a higher platelet count (1105 vs. 772 x 109/L, p = 0.025) and a mildly elevated red cell count (10.7 vs. 9.9 x 1012/L, p = 0.017) than mice transplanted with the control marrow. To begin to understand the MK signals responsible for HSC expansion in Pf4/FF1 mice, qRT-PCR of purified CD41+ MK cells identified up-regulation of FGF1 (8.1-fold, p = 0.02), Pf4 (8.0-fold, p = 0.0003), and CXCL12 (3.1-fold, p = 0.005) and down-regulation of TGFβ-1 (75%, p = 0.0005) in Pf4/FF1 mice compared to controls. Conclusions Pf4/FF1 mice developed an ET phenotype with thrombocytosis, splenomegaly and increased numbers of E-SLAM HSCs and CD41+ MK cells, despite JAK2V617F expression being restricted to MKs. Marrow histology suggested that both the osteoblastic niche and vascular niche might be affected by the JAK2V617F -mutant MKs. In addition, marrow cells from Pf4/FF1 mice repopulated wild-type recipients and induced a MPN phenotype. MKs from such mice displayed an altered pattern of cytokine expression, possibly contributing to the observed effects but the precise mechanism by which JAK2V617F-bearing MKs affect the MPN marrow niche and HSC expansion deserve further investigation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1995 ◽  
Vol 86 (3) ◽  
pp. 924-929 ◽  
Author(s):  
HS Ramshaw ◽  
SS Rao ◽  
RB Crittenden ◽  
SO Peters ◽  
HU Weier ◽  
...  

Abstract Bone marrow from animals treated with 5-fluorouracil (5FU) competes equally with normal marrow when assessed in vivo in an irradiated mouse, but shows markedly defective engraftment when transplanted into noncytoablated hosts. Using Southern Blot analysis and a Y-chromosome specific probe, we determined the level of engraftment of male donor cells in the bone marrow, spleen, and thymus of unprepared female hosts. We have confirmed the defective engraftment of marrow harvested 6 days after 5FU (FU-6) and transplanted into unprepared hosts and shown that this defect is transient; by 35 days after 5FU (FU-35), engraftment has returned to levels seen with normal marrow. FU-6 marrow represents an actively cycling population of stem cells, and we hypothesize that the cycle status of the stem cell may relate to its capacity to engraft in the nonirradiated host. Accordingly, we have evaluated the cycle status of engrafting normal and FU-6 marrow into normal hosts using an in vivo hydroxyurea technique. We have shown that those cells engrafting from normal marrow and over 70% of the cells engrafting from FU-6 marrow were quiescent, demonstrating no killing with hydroxyurea. We have also used fluorescent in situ hybridization (FISH) analysis with a Y-chromosome probe and demonstrated that normal and post-5FU engraftment patterns in peripheral blood were similar to those seen in bone marrow, spleen, and thymus. Altogether these data indicate that cells engrafting in normal, unprepared hosts are dormant, and the defect that occurs after 5FU is concomitant with the induction of these cells to transit the cell cycle.


Sign in / Sign up

Export Citation Format

Share Document