Transcriptional differences between JAK2-V617F and wild-type bone marrow cells in myeloproliferative neoplasm patients

Author(s):  
Debra Van Egeren ◽  
Baransel Kamaz ◽  
Shichen Liu ◽  
Maximilian Nguyen ◽  
Christopher R. Reilly ◽  
...  
Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4107-4107 ◽  
Author(s):  
Yohei Nakaya ◽  
Haruna Naito ◽  
Junko Homan ◽  
Seishi Sugahara ◽  
Tatsuya Horio ◽  
...  

Abstract Abstract 4107 A somatic point mutation of Janus Kinase 2 (JAK2) tyrosine kinase (JAK2 V617F) has been shown to occur at a high frequency in myeloproliferative neoplasm (MPN) patients. JAK2 V617F is a constitutively activated kinase that activates the JAK/STAT signaling pathway and dysregulates cell growth and function. These findings suggest that the inhibition of aberrant JAK2 activation has a therapeutic benefit. Our novel JAK2 inhibitor, NS-018, is highly active against JAK2 with an IC50 value of less than 1 nM, and it has 30–50-fold selectivities for JAK2 over other JAK-family kinases such as JAK1, JAK3 and Tyk2. We determined the X-ray structure of JAK2 in complex with NS-018. An Asp-Phe-Gly (DFG) motif is located at the N-terminus of the activation loop and regulates ATP binding. The resolved X-ray structure showed that NS-018 bound to JAK2 in the “DFG-in” active conformation. A molecular modeling study indicated that NS-018 would hardly bind to JAK2 in the “DFG-out” inactive conformation. In accordance with the structural analysis, NS-018 preferentially suppressed the growth of bone-marrow cells expressing activated JAK2. Thus, NS-018 reduced in a dose-dependent manner the number of erythroid colony-forming units (CFU-E) derived from bone-marrow cells taken from JAK2 V617F transgenic mice, but had only a limited effect on the number of colonies from wild-type mice (Figure A). NS-018 had no effect on the number of granulocyte-macrophage colony-forming units (CFU-GM) from either mouse strain. Furthermore, NS-018 showed potent antiproliferative activity against Ba/F3 cells expressing JAK2 V617F with an IC50 value of <100 nM but showed only minimal cytotoxicity against most other hematopoietic and non-hematopoietic cell lines (IC50 >3 μ M). In a mouse Ba/F3-JAK2 V617F leukemia model, NS-018 significantly prolonged survival during repeated oral administrations at 6.25 mg/kg bid and reduced splenomegaly at doses as low as 1.5 mg/kg bid. NS-018 was well tolerated at dosages of more than 100 mg/kg bid. In conclusion, NS-018 is a potent JAK2 inhibitor which preferentially inhibits an activated form of JAK2 and has potent in vitro and in vivo efficiency in preclinical studies. NS-018 is expected to be suitable for the treatment of MPN caused by aberrant JAK2 activation and its effectiveness will be verified by early-phase clinical investigations in the near future. JAK2 V617F preferential inhibition of erythrocyte colony growth Bone-marrow cells were collected from femurs of JAK2 V617F transgenic mice and same-strain BDF1 wild-type mice. (a) To detect CFU-E colonies, cells were treated with NS-018 in semisolid methylcellulose containing erythropoietin (EPO) and cell clusters were counted after incubation for two days. (b) To detect CFU-GM colonies, cells were treated with NS-018 in semisolid methylcellulose containing EPO, interleukin-3 (IL-3), IL-6 and stem cell factor and colonies were counted on day 7. Disclosures: Nakaya: Nippon Shinyaku Co., Ltd: Employment. Naito:Nippon Shinyaku Co., Ltd: Employment. Homan:Nippon Shinyaku Co., Ltd: Employment. Sugahara:Nippon Shinyaku Co., Ltd: Employment. Horio:Nippon Shinyaku Co., Ltd: Employment. Niwa:Nippon Shinyaku Co., Ltd: Employment. Shimoda:Nippon Shinyaku Co., Ltd: Research Funding.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2447-2447 ◽  
Author(s):  
Jing Zhang ◽  
Li Li ◽  
Alan D. Friedman ◽  
Donald Small ◽  
Ido Paz-Priel

Abstract Abstract 2447 Internal tandem duplication (ITD) of the fms-like tyrosine kinase 3 (FLT3) receptor is common in acute myeloid leukemia (AML) and is associated with a dismal outcome. Despite initial response, FLT3/ITD AMLs often relapse early, suggesting a residual population of resistant leukemia stem cells (LSC). Clinically, FLT3 inhibitors asmonotherapy have yet to improve outcome significantly and therefore, targeting additional pro-survival pathways may be necessary for this group of AML patients. Mice genetically egineered to express a hemizygous FLT3/ITD mutation develop a progressive, fatal, myeloproliferative neoplasm. Lin−cells isolated from the bone marrow of FLT3/ITD or control mice were subjected to gel shift analysis using a radio-labeled NF-kB binding site. This analysis demonstrated high levels of nuclear activation of NF-kB in the FLT3/ITD-expressing cells, suggesting its activation downstream of mutant FLT3 signaling. MV4–11 is a human AML-derived cell line harboring a homozygous FLT3/ITD mutation. Cells expressing high levels of aldehyde dehydrogenase (ALDH) have been shown to be enriched for LSC in primary AML samples and cell lines. High ALDH expressing MV4–11 cells were isolated using FACS and analyzed for NF-kB activation. Western blot analysis demonstrated preferential phosphorylation of NF-kB p65 by activated IKK on Ser536 in this subpopulation, compared with cells with low ALDH activity. These findings indicate activation of NF-kB in MV4–11 LSCs. We wanted to next test the requirement for NF-kB signaling in transformation by FLT3/ITD mutations. NF-kB p65 null mice die in utero. We therefore established C57BL/6 p65(flox/flox);Mx1-Cre mice. Intra-peritoneal injection of pIpC every other day for 7 doses efficiently deletes the RelA/p65 gene, resulting in expression of <1% of the corresponding RNA or protein. Despite effective excision of p65, the mice survive. Bone marrow cells harvested from control or p65(del/del) mice were transduced with a FLT3/ITD-expressing lentivirus and seeded in methylcellulose without cytokines. Equal transduction rate was verified by measurement of GFP expression by flow cytometry. Reproducibly, p65(del/del) marrow transduced with FLT3/ITD was ineffective in forming cytokine independent colonies, in contrast to wild-type marrow (5 +/− 0.6 vs. 55 +/− 6 colonies per 1E5 cells, P<0.001), and the few p65(del/del) colonies that resulted were smaller than those from p65 expressing wild-type marrow cells. Cells transduced with a lentiviral vector expressing GFP but not FLT3/ITD did not form colonies without cytokines, and p65(del/del) marrow formed normal numbers of colonies of normal size and distribution in the presence of IL-3, IL-6, and SCF. Sorafenib inhibits FLT3 signaling and kills MV4–11 cells with an IC50 of approximately 10 nM. Reproducibly, a sub-toxic dose of sorafenib (5 nM) combined with sub-toxic levels of the IKKb inhibitor IMD-0354 (400 nM) resulted in synergistic cell killing as indicated by the calculated combination index of 0.55. Currently, clinical efforts in FLT3/ITD leukemia concentrate on FLT3 inhibition alone. Our data suggest that canonical NF-kB may be an important pathway in FLT3/ITD AML and that simultaneously targeting FLT3 and NF-kB in this disease may be an effective approach. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3548-3548
Author(s):  
Joshua J. Oaks ◽  
Ramasamy Santhanam ◽  
Roger Briesewitz ◽  
Danilo Perrotti

Abstract Polycythemia vera (PV) is a myeloproliferative disorder (MPD) caused by a valine to phenylalanine mutation in the inhibitory JH2 “pseudokinase” domain of the signaling protein Janus Kinase 2 (Jak2). While Jak2 has several known regulators (e.g. SOCS1, SOCS3, PP2A, and SHP-1), the role played by these signaling molecules in the development of PV is still largely unclear. One of these regulators, the tumor suppressor phosphatase PP2A, has been found functionally inactivated in different hematologic myeloid and lymphoid malignancies characterized by the expression of constitutively activated oncogenic tyrosine kinases (e.g. BCR/ABL). To investigate the role of PP2A in the pathogenesis of Jak2 V617F+ MPDs and the potential therapeutic relevance of PP2A activating drugs (e.g. FTY720), we determined the effects of wild type and V617F Jak2 expression on PP2A activity and assessed the molecular and biological effects of FTY720 in hematopoietic precursor cell lines and/or primary lineage-negative bone marrow cells engineered to expressed either wild type or V617F Jak2 protein. Herein we report that PP2A activity is significantly reduced by about 82% and 78% (P&lt; 0.01) in mJak2 V617F-transduced growth factor-dependent and erythropoietin receptor-expressing 32Dcl3 (32D-EPO) and Ba/F3 cells, respectively, compared to MigR1-transduced controls. Furthermore, addition of the PP2A activator FTY720 (2.5μM) for 10 hours restored PP2A activity to 66% and 75% respectively compared to that of MigR1 controls. Mechanistically, we demonstrated that inactivation of PP2A was due to constitutive Jak2 activity. In fact, treatment of V617F Jak2-expressing cells with Jak inhibitor I (1μM; 10 hours) restored PP2A activity to 80% of controls in 32Dcl3 cells, while 600nM was sufficient to restore activity to 108% of controls in Ba/f3 cells. Likewise, transduction of murine lineage-negative bone marrow cells with wild-type Jak2 produced a 62% reduction in PP2A activity (P&lt;0.01), while expression of the Jak2 V617F mutant produced a 97% reduction (P&lt; 0.01). Moreover, as we previously reported for BCR/ABL, pharmacologic restoration of PP2A activity by treatment of Ba/F3 or 32D-EPO cells with the PP2A activator FTY720 (2.5μM for 10 hours) led to reduced expression and dephosphorylation of wild type and V617F Jak2. This, in turn, resulted in a 60% reduction in the colony forming ability of IL-3 cultured cells overexpressing wild type Jak2, while a 94% suppression of colony formation was evident in Jak2 V617-expressing cells maintained in the absence of cytokines. Moreover, addition of IL-3 to the FTY720-containing semisolid medium restored viability to levels similar to those of FTY720-treated wild type Jak2-expressing cells (70% suppression of colony formation upon treatment), indicating that restoration of PP2A activity counteracts cytokine-independent pathways triggered by the V617F Jak2 mutated tyrosine kinase. Altogether, these results suggest that inactivation of PP2A is essential for Jak2 (V617F mutant included) -driven cell/proliferation and survival and that pharmacologic activation of PP2A might represent a potential avenue for treatment of PV and, perhaps, of other MPDs characterized by the expression of a mutated Jak2 kinase.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2531-2531
Author(s):  
Kotaro Shide ◽  
Haruko K. Shimoda ◽  
Takashi Kumano ◽  
Kennosuke Karube ◽  
Takuro Kameda ◽  
...  

Abstract An acquired JAK2 V617F mutation has been detected in up to 90% of patients with polycythemia vera (PV) and in a sizeable proportion of patients with other myeloproliferative disorders such as essential thrombocythemia (ET) and idiopathic myelofibrosis (IMF). However, how a single mutation may be responsible for such different clinical phenotypes is unknown. Mice transplanted with bone marrow cells in which V617F JAK2 was retrovirally expressed developed PV-like features, but not ET or IMF. To address the contribution of this mutation to the pathogenesis of these three MPDs, we generated transgenic mice expressing V617F JAK2 driven by the murine H2Kb promoter. We established two lines. The expression of V617F JAK2 mRNA in bone marrow cells was 0.45 and 1.35 that of endogenous wild-type JAK2 in the two lines. One line showed leukocytosis after 4 months of age, with a predominance of granulocytes. Among 43 mice, examined after 3 months of age, 8 (19%) showed polycythemia and 14 (33%) showed thrombocythemia. Two polycythemia cases also showed thromobocytosis. The other line showed extreme leukocytosis and thromobocytosis at one month of age. The leukocytosis progressed as the animals aged, but the thrombocytosis tended to resolve at 8 months. They showed anemia that means Hb value from 9 to 10 g/dL at one month old. Myeloid cells and megakaryocytes were predominant in the bone marrow of these animals, and splenomegaly with myeloid cell and megakaryocyte invasion was observed. We conclude that in vivo expression of V617F JAK2 results in ET-like, IMF-like, and PV-like disease.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3897-3897 ◽  
Author(s):  
Kotaro Shide ◽  
Takuro Kameda ◽  
Vadim Markovtsov ◽  
Takuya Matsunaga ◽  
Haruko Shimoda ◽  
...  

Abstract Abstract 3897 Poster Board III-833 The V617F mutation of JAK2 (JAK2-V617F) occurs at a high frequency in Philadelphia chromosome-negative myeloproliferative diseases (MPDs). JAK2-V617F leads to constitutive activation of the JAK2 kinase, causing cytokine independent growth in cell lines and development of MPD in mice. These data suggest that aberrant activation of JAK2 plays a pivotal role in the pathophysiology of MPDs and targeting JAK2-V617F may be therapeutically useful. Some orally bioavailable inhibitors of JAK2 are already in clinical trials. R723 is a potent and highly selective (in vitro kinase assay: JAK2 IC50=2nM, JAK1 IC50 >1μM, JAK3 IC50 =24nM) small molecule inhibitor of JAK2. We tested R723 in a murine model of MPD induced by JAK2-V617F. H2Kb promoter-controlled JAK2-V617F expressing mice (V617F-TG) show extreme leukocytosis, thrombocytosis and progressive anemia. They also show hepato-splenomegaly with extramedullary hematopoiesis. Megakaryocytes are predominant in bone marrow and new formation of bony trabeculae and accumulation of fibers are seen. In vitro analysis of bone marrow cells shows constitutive activation of STAT5 and formation of cytokine-independent growth of erythroid colony-forming units (CFU-E). They exhibit high mortality compared to wild-type controls. At first, we assessed the effect of R723 on wild-type (WT) and V617F-TG bone marrow cells in vitro. R723 inhibited cytokine-independent CFU-E growth and constitutive activation of STAT5 of V617F-TG cells. R723 also inhibited cytokine-dependent colony growth and cytokine-induced STAT5 activation in both WT and V617F-TG cells at the same level. Next, we assessed the effect of R723 in vivo. On development of MPD, V617F-TG mice were divided into treatment or vehicle control groups. R723 was administered by oral gavage at 35mg/kg or 70mg/kg bid for 16 weeks, whereas the control groups received vehicle only. Mice were followed by blood counts and a subset of mice was euthanized for detailed histopathology and FACS analysis. In treated mice, there was a significant reduction in leukocyte count in both groups, and a reduction in platelet count in the high dose group. There was no improvement in anemia. They showed a dose-dependent-reduction of hepato-splenomegaly. Histopathology and FACS analysis revealed a reduction of myeloid cells, with partially restored architecture in spleen. In bone marrow, R723 had little effect on progression of fibrosis and megakaryocyte hyperplasia. During the time course of study, 6 out of 23 mice died in the vehicle group, whereas 1 out of 26 mice died in the 70mg/kg group. There was a statistically significant prolongation of survival in the 70mg/kg group (p<0.05). R723 shows therapeutic efficacy in a murine model of MPD induced by JAK2-V617F, and could become the basis for a next generation of potent and selective compounds targeting JAK2-dependent MPDs. Disclosures: Markovtsov: Rigel Pharmaceuticals,Inc.: Employment. Bhamidipati:Rigel Pharmaceuticals,Inc.: Employment. Park:Rigel Pharmaceuticals,Inc.: Employment. Torneros:Rigel Pharmaceuticals,Inc.: Employment. Duan:Rigel Pharmaceuticals,Inc.: Employment. Hitoshi:Rigel Pharmaceuticals,Inc.: Employment. Shimoda:Rigel Pharmaceuticals,Inc.: Research Funding.


1998 ◽  
Vol 331 (3) ◽  
pp. 733-742 ◽  
Author(s):  
Masafumi YOSHIMURA ◽  
Yoshito IHARA ◽  
Tetsuo NISHIURA ◽  
Yu OKAJIMA ◽  
Megumu OGAWA ◽  
...  

Several sugar structures have been reported to be necessary for haemopoiesis. We analysed the haematological phenotypes of transgenic mice expressing β-1,4 N-acetylglucosaminyltransferase III (GnT-III), which forms bisecting N-acetylglucosamine on asparagine-linked oligosaccharides. In the transgenic mice, the GnT-III activity was elevated in bone marrow, spleen and peripheral blood and in isolated mononuclear cells from these tissues, whereas no activity was found in these tissues of wild-type mice. Stromal cells after long-term cultures of transgenic-derived bone marrow and spleen cells also showed elevated GnT-III activity, compared with an undetectable activity in wild-type stromal cells. As judged by HPLC analysis, lectin blotting and lectin cytotoxicity assay, bisecting GlcNAc residues were increased on both blood cells and stromal cells from bone marrow and spleen in transgenic mice. The transgenic mice displayed spleen atrophy, hypocellular bone marrow and pancytopenia. Bone marrow cells and spleen cells from transgenic mice produced fewer haemopoietic colonies. After lethal irradiation followed by bone marrow transplantation, transgenic recipient mice showed pancytopenia compared with wild-type recipient mice. Bone marrow cells from transgenic donors gave haematological reconstitution at the same level as wild-type donor cells. In addition, non-adherent cell production was decreased in long-term bone marrow cell cultures of transgenic mice. Collectively these results indicate that the stroma-supported haemopoiesis is compromised in transgenic mice expressing GnT-III, providing the first demonstration that the N-glycans have some significant roles in stroma-dependent haemopoiesis.


Blood ◽  
1997 ◽  
Vol 90 (6) ◽  
pp. 2148-2159 ◽  
Author(s):  
Harshal H. Nandurkar ◽  
Lorraine Robb ◽  
David Tarlinton ◽  
Louise Barnett ◽  
Frank Köntgen ◽  
...  

Abstract Interleukin-11 (IL-11) is a pleiotropic growth factor with a prominent effect on megakaryopoiesis and thrombopoiesis. The receptor for IL-11 is a heterodimer of the signal transduction unit gp130 and a specific receptor component, the α-chain (IL-11Rα). Two genes potentially encode the IL-11Rα: the IL11Ra and IL11Ra2 genes. The IL11Ra gene is widely expressed in hematopoietic and other organs, whereas the IL11Ra2 gene is restricted to only some strains of mice and its expression is confined to testis, lymph node, and thymus. To investigate the essential actions mediated by the IL-11Rα, we have generated mice with a null mutation of IL11Ra (IL11Ra−/−) by gene targeting. Analysis of IL11Ra expression by Northern blot and reverse transcriptase-polymerase chain reaction, as well as the absence of response of IL11Ra−/− bone marrow cells to IL-11 in hematopoietic assays, further confirmed the null mutation. Compensatory expression of the IL11Ra2 in bone marrow cells was not detected. IL11Ra−/− mice were healthy with normal numbers of peripheral blood white blood cells, hematocrit, and platelets. Bone marrow and spleen contained normal numbers of cells of all hematopoietic lineages, including megakaryocytes. Clonal cultures did not identify any perturbation of granulocyte-macrophage (GM), erythroid, or megakaryocyte progenitors. The number of day-12 colony-forming unit-spleen progenitors were similar in wild-type and IL11Ra−/− mice. The kinetics of recovery of peripheral blood white blood cells, platelets, and bone marrow GM progenitors after treatment with 5-flurouracil were the same in IL11Ra−/− and wild-type mice. Acute hemolytic stress was induced by phenylhydrazine and resulted in a 50% decrease in hematocrit. The recovery of hematocrit was comparable in IL11Ra−/− and wild-type mice. These observations indicate that IL-11 receptor signalling is dispensable for adult hematopoiesis.


Blood ◽  
1997 ◽  
Vol 89 (1) ◽  
pp. 41-48 ◽  
Author(s):  
Helga Björgvinsdóttir ◽  
Chunjin Ding ◽  
Nancy Pech ◽  
Mary A. Gifford ◽  
Ling Lin Li ◽  
...  

Abstract The X-linked form of chronic granulomatous disease (X-CGD), an inherited deficiency of the respiratory burst oxidase, results from mutations in the X-linked gene for gp91phox, the larger subunit of the oxidase cytochrome b. The goal of this study was to evaluate the impact of retroviral-mediated gene transfer of gp91phox on host defense against Aspergillus fumigatus in a murine model of X-CGD. Retrovirus vectors constructed using the murine stem cell virus (MSCV) backbone were used for gene transfer of the gp91phox cDNA into murine X-CGD bone marrow cells. Transduced cells were transplanted into lethally irradiated syngeneic X-CGD mice. After hematologic recovery, superoxide production, as monitored by the nitroblue tetrazolium (NBT) test, was detected in up to ≈80% of peripheral blood neutrophils for at least 28 to 35 weeks after transplantation. Neutrophil expression of recombinant gp91phox and superoxide production were significantly less than wild-type neutrophils. However, 9 of 9 mice with ≈50% to 80% NBT+ neutrophils after gene transfer did not develop lung disease after respiratory challenge with 150 to 500 A fumigatus spores, doses that produced disease in 16 of 16 control X-CGD mice. In X-CGD mice transplanted with mixtures of wild-type and X-CGD bone marrow, ≥5% wild-type neutrophils were required for protection against A fumigatus challenge. These data suggest that expression of even low levels of recombinant gp91phox can substantially improve phagocyte function in X-CGD, although correction of very small percentage of phagocytes may not be sufficient for protection against A fumigatus.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 460-460 ◽  
Author(s):  
Charlotte Gruber ◽  
Rebekka Grundler ◽  
Georg Haecker ◽  
Falco Fend ◽  
Christian Peschel ◽  
...  

Abstract Anaplastic large cell lymphomas (ALCLs) define a subgroup of aggressive non-Hodgkin’s lymphomas. In 40–60% of systemic ALCLs, a t(2;5) (p23;q35) translocation is found, which generates a fusion gene between nucleophosmin (NPM) and the receptor tyrosine kinase gene ALK (anaplastic lymphoma kinase). The NPM-ALK chimeric gene encodes a constitutively activated tyrosine kinase and is believed to initiate the process of lymphomagenesis. Since NPM-ALK has been shown to activate PI3-kinase and STAT3, proteins that are involved in apoptosis regulation, altered apoptosis might contribute to ALCL development. Bcl-3, which exerts an anti-apoptotic effect in B and T lymphocytes, has been reported to be up regulated in a subgroup of ALCLs. This raised the question whether Bcl-3 is required for tumour development induced by NPM-ALK. In a first set of experiments, the expression levels of Bcl-3 in NPM-ALK negative and NPM-ALK positive tumour cells were analysed. Bcl-3 expression was enhanced in NPM-ALK expressing cells compared to NPM-ALK negative cells or cells expressing a kinase dead NPM-ALK mutant. Next we used primary murine bone marrow cells from wt versus Bcl-3 −/− mice in order to clarify the contribution of Bcl-3 to NPM-ALK induced transformation. We employed a retroviral infection system utilizing a MCSV-based vector co-expressing NPM-ALK together with the enhanced green fluorescent protein (EGFP) via an internal ribosomal entry site to infect Bcl-3−/− and wild type (wt) bone marrow cells. Transformation of bone marrow cells was analysed by methylcellulose assay without cytokines. Transformation of NPM-ALK infected Bcl-3−/− bone marrow cells was comparable to NPM-ALK infected BL6 wt bone marrow cells. No colony formation was detectable after transfection of Bcl-3−/− bone marrow with empty vector as control. Finally, we utilized a murine transplantation model of ALCL. Lethally irradiated BL6 wt mice were transplanted with retrovirally NPM-ALK infected wild type or Bcl-3−/− bone marrow cells. As a control, BL6 wt mice received a transplant of Bcl-3−/− bone marrow cells infected with supernatant from viral producer cells transfected with empty vector. Mice transplanted with NPM-ALK infected wild type or Bcl-3−/− bone marrow cells rapidly died within a median survival time of 16 and 17 days respectively, whereas mice transplanted with Bcl-3 −/− bone marrow cells transfected with empty vector survived healthy for more than 300 days. Diseased mice macroscopically showed involvement of the spleen, predominantly. Histologically, spleens of diseased mice showed an extensive infiltration of ALK-positive tumour cells with proliferation of large histiocytic cells both in mice transplanted with NPM-ALK infected Bcl-3−/− and wild type bone marrow. In both groups, FACS-analysis revealed a high percentage of EGFP positive and thus NPM-ALK positive cells in bone marrow and spleen. In conclusion, NPM-ALK is able to transform bone marrow cells and to induce a lymphoma-like disease in the absence of Bcl-3. Bcl-3 is thus dispensable for ALCL development in a murine mouse model. Lack of Bcl-3 in the knockout mice may be compensated by the expression of other proteins. Therefore, Bcl-3 upregulation in a subgroup of human ALCLs may be not critical for lymphoma development.


Sign in / Sign up

Export Citation Format

Share Document