Inhibition of JAK2 V617F-Induced Erythroid Skewing of Hematopoietic Stem Cell Differentiation with a Selective JAK2 Antagonist.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3616-3616 ◽  
Author(s):  
Ifat Geron ◽  
Charlene Barroga ◽  
Jason Gotlib ◽  
Edward Kavalerchik ◽  
Annelie Abrahamsson ◽  
...  

Abstract Introduction Polycythemia vera (PV) is characterized by excessive production of erythroid cells and in most cases a point mutation (V617F) in the Jak2 cytokine signaling kinase. We investigated whether a selective JAK2 inhibitor decreased Jak2 V617F induced erythroid differentiation. Methods Wild-type and mutant Jak2 V617F genes were excised from the retroviral Jak2-mus-MSCV-neo vector (Levine et al), cloned into the lentiviral vector pLV CMV IRES2 GFP and their presence verified by DNA sequencing. Lentiviral vectors bearing the wild-type and mutant Jak2 genes or vector alone were used to transduce human peripheral blood CD34+ cells, which were then divided for plating into megacult medium for megakaryocytic colony growth and methylcellulose culture for enumeration of all other progenitor cell types. Normal cord blood HSC (CD34+/CD38−/CD90+) were clone sorted with the FACS Aria and transduced with no vector, backbone vector, wild-type JAK2 or mutant JAK2 vector in methocult media (Stem Cell Technologies Inc, GF+ H4435) +/− 300 nM of a selective JAK2 inhibitor, TG101348. Colonies were scored at day 14. RNA was isolated from the colonies (Qiagen RNeasy kit) and RT-PCR was performed with wild-type and mutant JAK2 allele specific primers. Results Transduction of cord blood HSC with the mutant Jak2 vector resulted in skewed erythroid colony formation compared to wild-type Jak2, vector alone and untransduced HSC (Figure 1; n=3). RT-PCR with murine Jak2 specific primers resulted in ~900 bp fragments corresponding to murine Jak2 from colonies transduced with the wild-type and mutant Jak2 and confirmed by sequencing, but not those from colonies transduced with the vector alone or the untransduced cells. Like the results in cord blood cells, adult peripheral blood CD34+ cells transduced with the mutant Jak2 developed a skewed developmental pattern, with far greater erythroid colony formation compared to wild-type Jak2 or vector alone. In megacult assays, CD34+ cells transduced with the mutant Jak2 had similar megakaryocytic potential as wild-type Jak2 or vector alone. Addition of TG101348 (300 nM), inhibited mutant kinase-induced erythroid colony formation (Figure 1) in 3 experiments while 100– 300 nM was inhibitory to PV (n=2 patients) HSC and progenitors. Current experiments focus on inhibition of Jak2 in a bioluminescent highly immunocompromised mouse model of Jak2V617F-induced myeloproliferation (Figure 2). Conclusion JAK2 V617F skews differentiation of HSC toward the erythroid lineage and may be inhibited with a selective JAK2 inhibitor - TG101348. Figure 1. In vitro JAK2 Inhibition. Figure 1. In vitro JAK2 Inhibition. Figure 2. Bioluminescent JAK2 V617F-induced Myeloproliferation Model. Figure 2. Bioluminescent JAK2 V617F-induced Myeloproliferation Model.

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 ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 794-794
Author(s):  
Fan Liu ◽  
Xinyang Zhao ◽  
Fabiana Perna ◽  
Lan Wang ◽  
Priya Koppikar ◽  
...  

Abstract Abstract 794 Background: The cytoplasmic, non-receptor JAK2 tyrosine kinase is mutated at amino acid residue 617 (from valine to phenylalanine) in most patients with myeloproliferative neoplasms (MPNs), resulting in a constitutively activated kinase that phosphorylates STAT proteins in the absence of upstream signals. Overexpression of JAK2V617F leads to cytokine-independent growth of Ba/F3 cells and the JAK2V617F transgenic and knockin mice develop a disease phenotype resembling human polycythemia vera. Results: We hypothesized that the JAK2V617F occurs so consistently in MPNs because it gains some functional property. The type II arginine methyltransferase PRMT5 was initially identified because of its interaction with JAK2 in a yeast two hybrid screen. We examined the interaction between JAK2 and PRMT5 and found that JAK2V617F and JAK2K539L (another active JAK2 kinase) bound PRMT5 more strongly than did wild-type JAK2. PRMT5 mediates the symmetrical dimethylation of arginine residues within histones H2A and H4 and methylates other cellular proteins as well, such as p53. The oncogenic forms of JAK2 acquire the ability to phosphorylate PRMT5, which greatly impaired its methyltransferase activity. We have shown the in vivo importance of this post-translation modification as treating JAK2V617F-positive cells (but not the wild-type JAK2-harboring cells) with different JAK2 inhibitors significantly increased histone arginine methylation levels. To define the effect of inhibiting PRMT5 activity on hematopoiesis, we knocked down PRMT5 in human cord blood derived CD34+ cells using shRNA and observed increased colony formation and erythroid differentiation; In contrast, PRMT5 overexpression in these cells led to reduced colony formation and inhibition of erythroid differentiation. Furthermore, overexpression of PRMT5, especially a phosphorylation site mutant form of PRMT5 (PRMT5M6), diminishes the proliferative and erythroid generating capacity of JAK2V617F+ CD34+ cells isolated from MPN patients to a greater degree than normal cord blood CD34+ cells. Importantly, we found marked increase in PRMT5 phosphorylation in JAK2V617F-positive MPN patents relative to normal cord blood CD34+ cells, suggesting that this phosphorylation is important for the myeloproliferation phenotype. Conclusion: we show that the oncogenic mutant forms of JAK2 kinase, such as JAK2V617F and JAK2K539L, are not simply constitutively active forms of wild-type JAK2, rather they have specific gains-of-function that allow them to phosphorylate PRMT5 and down-regulate its enzymatic activity. Inhibition of PRMT5 contributes to the myeloproliferation and erythroid differentiation promoting effects of JAK2V617F. This gain-of –function mutation results in cross-talk between oncogenic kinases and histone arginine methylation. Taken together, we demonstrate a novel link between the mutant JAK2 kinases and PRMT5 methyltransferase activity, which contributes to MPN pathogenesis. Further insights about the shared gene expression profile of JAK2 inhibition vs. PRMT5 knockdown will be presented to understand the basics for the behavior change in hematopoietic stem/progenitor cells brought about by these two interventions. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1433-1433
Author(s):  
Edward Kavalerchik ◽  
Charlene Barroga ◽  
Larissa Balaian ◽  
Jennifer M. Black ◽  
Catriona H.M. Jamieson

Abstract Abstract 1433 Poster Board I-456 Introduction Many myeloproliferative neoplasms (MPN) arise in the hematopoietic stem cell (HSC) population upon acquisition of a somatic mutation leading to an aberrantly activated tyrosine kinase, JAK2 V617F. Identification of cell type and context specific genetic and epigenetic events driving leukemic transformation of MPNs has been hampered by the limited supply of the MPN patient blood and marrow samples. Recent publications demonstrate that human embryonic stem cells (hESC) can be differentiated to HSCs efficiently upon coculture with stromal cell lines derived from the murine aorta-gonad-mesonephros (AGM) region. These methods can provide a potentially limitless supply of self-renewing stem cells. We have utilized two hES cell lines, H9 and Hues9 cells, to differentiate toward HSC. We developed novel in vitro and in vivo model systems involving lentivirally enforced expression of genes, such as JAK2V617F, wild type JAK2 and/or activated beta-catenin, in an attempt to generate MPN stem cells. Methods and Results To generate MPN stem cells in initial experiments (n=4), karyotypically normal Hues9 cells were lentivirally transduced with luciferase-GFP and constitutively active beta-catenin (Beta-Cat) alone or in combination with 1) JAK2 V617F, 2) wild-type JAK2, or 3) backbone lentiviral vector. The engraftment capacity of these genetically modified Hues9 cells was compared following intrahepatic transplantation (105-106 cells/mouse) of neonatal RAG2-/-gamma c-/- mice. Lentiviral transduction efficiency was 50-60%. Bioluminescent imaging demonstrated human engraftment at 6 and 11 weeks in all subgroups. The greatest proportion of human cells was noted in the thymus at week 6 and 11, and in the spleens at week 6. There was a 2.4 fold increase in thymic engraftment of CD34+CD45+ cells derived from JAK2 V617F transduced Hues9 cells and a 1.7-fold increase in CD38+CD45+ cells in the JAK2-wild type subset of mice compared with backbone controls. However, overall engraftment rates were low. To determine if hESC differentiation into CD34+ cells improved engraftment, both H9 and Hues9 hES cells were plated in 6 well plates onto mitomycin treated stromal cell lines AM20.1B4 (AM) and UG26.1B6 (UG) that were derived from aorta/mesenchyme and the urogenital ridge of the AGM region of 10 dpc murine embryos respectively. Peak CD34 expression was observed on day 10-12 (n=4) with a sharp decline in CD34 expression by day 18 (n=2). Notably, CD34+ cells began to express CD31 by day 18 with over 50% of CD34+ cells expressing CD31 by day 21 (n=2). Both undifferentiated H9 and Hues9 hES cells as well as CD34+ derivatives expressed CD90. RUNX-1 and GATA-1 expression was higher on day 12 compared to day 18 by Q-RT-PCR in H9 cultured on UG stroma. Plating of 100,000 hESC from day 10-12 cultures with UG stroma generated colonies in hematopoietic progenitor assays with in vitro replating potential while day 18-24 hESC cultures did not. In vitro derived CD34+ cells from day 13 H9 cultures with UG stroma were double selected for CD34 using immunomagnetic beads. The cells were transduced for 6 hr with lentiviral luciferase GFP, Beta-Cat alone (N=4), or Beta-Cat and lentiviral backbone vector (LV) (N=4), wild-type JAK2 (N=4) or JAK2 V617F. The cells were transplanted intrahepatically (5×104 cells/mouse) as above and monitored for engraftment by bioluminescent imaging. Human engraftment was detected at 6 weeks in 2 out of 3 JAK2 V617F/Beta-Cat, 1 of 4 wild-type JAK2/ Beta-Cat, and 0 of 4 Beta-Cat transplanted mice. Conclusions Co-culturing of hES cell lines, H9 and Hues9, on AGM stromal cell lines appears to generate replatable colonies in hematopoietic progenitor assays. Furthermore, engraftment of lentiviral luciferase transduced hESC in RAG2-/-gamma c-/- mouse hematopoietic organs can be monitored by non-invasive bioluminescent imaging. Although enhanced by lentivirally enforced expression of genes that promote hematopoietic differentiation, hematopoietic engraftment is not robust suggesting a relative dearth of environmental cues that promote human hematopoietic differentiation. Transplantation of genetically engineered hESC together with supportive stroma as well as secondary transplantation will be used to monitor changes in hESC derived HSC self-renewal capacity upon introduction of oncogenes in order to provide further insights into MPN stem cell generation. Disclosures Barroga: Wintherix: Employment. Jamieson: Merck: Research Funding; Pfizer: Research Funding; Wintherix: Consultancy; TargeGen: Research Funding; Celgene: Research Funding; Coronado Biosciences: Research Funding.


Blood ◽  
2008 ◽  
Vol 111 (12) ◽  
pp. 5663-5671 ◽  
Author(s):  
Elizabeth O. Hexner ◽  
Cynthia Serdikoff ◽  
Mahfuza Jan ◽  
Cezary R. Swider ◽  
Candy Robinson ◽  
...  

Abstract Recent studies have demonstrated that patients with myeloproliferative disorders (MPDs) frequently have acquired activating mutations in the JAK2 tyrosine kinase. A multikinase screen determined that lestaurtinib (formerly known as CEP-701) inhibits wild type JAK2 kinase activity with a concentration that inhibits response by 50% (IC50) of 1 nM in vitro. We hypothesized that lestaurtinib would inhibit mutant JAK2 kinase activity and suppress the growth of cells from patients with MPDs. We found that lestaurtinib inhibits the growth of HEL92.1.7 cells, which are dependent on mutant JAK2 activity for growth in vitro and in xenograft models. Erythroid cells expanded from primary CD34+ cells from patients with MPDs were inhibited by lestaurtinib at concentrations of 100 nM or more in 15 of 18 subjects, with concomitant inhibition of phosphorylation of STAT5 and other downstream effectors of JAK2. By contrast, growth of erythroid cells derived from 3 healthy controls was not significantly inhibited. These results demonstrate that lestaurtinib, in clinically achievable concentrations, inhibits proliferation and JAK2/STAT5 signaling in cells from patients with MPDs, and therefore holds promise as a therapeutic agent for patients with these disorders.


Transfusion ◽  
2008 ◽  
Vol 48 (10) ◽  
pp. 2235-2245 ◽  
Author(s):  
Eun Jung Baek ◽  
Han-Soo Kim ◽  
Sinyoung Kim ◽  
Honglien Jin ◽  
Tae-Yeal Choi ◽  
...  

Blood ◽  
2009 ◽  
Vol 113 (2) ◽  
pp. 458-461 ◽  
Author(s):  
Shinji Kunishima ◽  
Ryoji Kobayashi ◽  
Tomohiko J. Itoh ◽  
Motohiro Hamaguchi ◽  
Hidehiko Saito

Abstract Congenital macrothrombocytopenia is a genetically heterogeneous group of rare disorders. We identified the first TUBB1 mutation, R318W, in a patient with congenital macrothrombocytopenia. The patient was heterozygous for Q43P, but this single-nucleotide polymorphism (SNP) did not relate to macrothrombocytopenia. Although no abnormal platelet β1-tubulin localization/marginal band organization was observed, the level of β1-tubulin was decreased by approximately 50% compared with healthy controls. Large and irregular bleb protrusions observed in megakaryocytes derived from the patient's peripheral blood CD34+ cells suggested impaired megakaryocyte fragmentation and release of large platelets. In vitro transfection experiments in Chinese hamster ovary (CHO) cells demonstrated no incorporation of mutant β1-tubulin into microtubules, but the formation of punctuated insoluble aggregates. These results suggested that mutant protein is prone to aggregation but is unstable within megakaryocytes/platelets. Alternatively, mutant β1-tubulin may not be transported from the megakaryocytes into platelets. W318 β1-tubulin may interfere with normal platelet production, resulting in macrothrombocytopenia.


FEBS Letters ◽  
1998 ◽  
Vol 441 (1) ◽  
pp. 39-42 ◽  
Author(s):  
Etsuko Satoh ◽  
Hideyo Hirai ◽  
Tohru Inaba ◽  
Chihiro Shimazaki ◽  
Masao Nakagawa ◽  
...  

2021 ◽  
Author(s):  
Lei Xu ◽  
Zhan Gao ◽  
Zhou Yang ◽  
Mingyi Qu ◽  
Huilin Li ◽  
...  

Blood ◽  
2000 ◽  
Vol 96 (13) ◽  
pp. 4169-4177 ◽  
Author(s):  
Adeline Lepage ◽  
Marylène Leboeuf ◽  
Jean-Pierre Cazenave ◽  
Corinne de la Salle ◽  
François Lanza ◽  
...  

Abstract Megakaryocytopoiesis is a complex multistep process involving cell division, endoreplication, and maturation and resulting in the release of platelets into the blood circulation. Megakaryocytes (MK) progressively express lineage-restricted proteins, some of which play essential roles in platelet physiology. Glycoprotein (GP)Ib-V-IX (CD42) and GPIIb (CD41) are examples of MK-specific proteins having receptor properties essential for platelet adhesion and aggregation. This study defined the progressive expression of the GPIb-V-IX complex during in vitro MK maturation and compared it to that of GPIIb, an early MK marker. Human cord blood CD34+ progenitor cells were cultured in the presence of cytokines inducing megakaryocytic differentiation. GPIb-V-IX expression appeared at day 3 of culture and was strictly dependent on MK cytokine induction, whereas GPIIb was already present in immature CD34+ cells. Analysis by flow cytometry and of the messenger RNA level both showed that GPV appeared 1 day later than GPIb-IX. Microscopy studies confirmed the late appearance of GPV, which was principally localized in the cytoplasm when GPIb-IX was found on the cell surface, suggesting a delayed program of GPV synthesis and trafficking. Cell sorting studies revealed that the CD41+GPV+ population contained 4N and 8N cells at day 7, and was less effective than CD41+GPV− cells in generating burst-forming units of erythrocytes or MK colonies. This study shows that the subunits of the GPIb-V-IX complex represent unique surface markers of MK maturation. The genes coding for GPIb-IX and GPV are useful tools to study megakaryocytopoiesis and for tissue-specific or conditional expression in mature MK and platelets.


Sign in / Sign up

Export Citation Format

Share Document