In Vitro Expansion of Polycythemia Vera Progenitors Favors Expansion of Erythroid Precursors without JAK2 V617F Mutation.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3506-3506 ◽  
Author(s):  
Josef T. Prchal ◽  
Ko-Tung Chang ◽  
Jaroslav Jelinek ◽  
Yongli Guan ◽  
Amos Gaikwad ◽  
...  

Abstract A single acquired point mutation of JAK2 1849G>T (V617F), a tyrosine kinase with a key role in signal transduction from growth factor receptors, is found in 70%–97% of patients with polycythemia vera (PV). In the studies of tyrosine kinase inhibitors on JAK2 1849G>T (see Gaikwad et all abstract at this meeting) we decided to study the possible therapeutic effect of these agents using native in vitro expanded cells from peripheral blood. To our surprise, the in vitro expansion of PV progenitors preferentially augmented cells without JAK2 1849G>T mutation. We used a 3 step procedure to amplify erythroid precursors in different stages of differentiation from the peripheral blood of 5 PV patients previously found to be homozygous or heterozygous for the JAK2 1849G>T mutation. In the first step (days 1–7), 106/ml MNCs were cultured in the presence of Flt-3 (50 ng/ml), Tpo (100 ng/ml), and SCF (100 ng/ml). In the second step (days 8–14), the cells obtained on day 7 were re-suspended at 106/ml in the same medium with SCF (50 ng/ml), IGF-1 (50 ng/ml), and 3 units/ml Epo. In the third step, the cells collected on day 14 were re-suspended at 106/ml and cultured for two more days in the presence of the same cytokine mixture as in the step 2 but without SCF. The cultures were incubated at 37oC in 5% CO2/95% air atmosphere and the medium renewed every three days to ensure good cell proliferation. The expanded cells were stained with phycoerythrin-conjugated anti-CD235A (glycophorin) and fluorescein isothiocyanate-conjugated anti-human-CD71 (transferrin receptor) monoclonal antibodies and analyzed by flow cytometry. The cells were divided by their differential expression of these antigens into 5 subgroups ranging from primitive erythroid progenitors (BFU-Es and CFU-Es) to polychromatophilic and orthochromatophilic erythroblasts; over 70% of harvested cells were early and late basophilic erythroblasts. The proportion of JAK2 1849G>T mutation in clonal PV granulocytes (GNC) before in vitro expansion and in expanded erythroid precursors was quantitated by pyrosequencing (Jelinek, Blood in press) and is depicted in the Table. These data indicate that in vitro expansion of PV progenitors favors expansion of erythroid precursors without JAK2 V617F mutation. Since three PV samples were from females with clonal granulocytes, erythrocytes, and platelets, experiments were underway to determine if the in vitro expanded erythroid cells were clonal PV cells without JAK2 V617F mutation, or derived from polyclonal rare circulating normal hematopoietic progenitors. The Proportion of JAK2 T Allele Patients GNC T Allele (%) Expanded Cells T Allele (%) PV1 (Female) 81 10 PV2 (Male) 77 28 PV3 (Male) 44 42 PV4 (Female) 78 19 PV5 (Female) 78 28

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 5054-5054 ◽  
Author(s):  
Lourdes Florensa ◽  
Beatriz Bellosillo ◽  
Leonor Arenillas ◽  
Liandong Ma ◽  
Richard Walgren ◽  
...  

Abstract Abstract 5054 Introduction: The discovery of JAK2 V617F mutation in patients with myeloproliferative disorders (MPD) has opened new perspectives for the development of targeted therapies. We have studied the efficacy of a novel molecule LY2784544 with JAK2 inhibitory activity in the in vitro growth of myeloid progenitors from JAK2 V617F-positive polycythemia vera (PV) patients. Objectives: To investigate the efficacy of LY2784544 in the inhibition of endogenous(e)BFU-E and CFU-GM growth in PV patients. Methods: In vitro cultures in semisolid media were performed from peripheral blood mononuclear cells (PBMC) of 6 PV patients who had never received cytoreductive treatment (4 patients with homozygous JAK2 V617F and 2 patients with heterozygous JAK2 V617F). PBMC were suspended in methylcellulose (Methocult. StemCell, Vancouver, Canada) without the addition of EPO and containing 0–30.0 μM LY2784544 drug. Concurrent plates containing EPO were plated as control cultures. The medium was distributed in multidishes and they were incubated at 37° with 5% CO2 and 95% humidity. Hemoglobinized colonies and granulomonocytic colonies were counted on day 14 by standard criteria (BFU-E defined by an aggregate of >50 hemoglobinized cells or three or more erythroid subcolonies and CFU-GM was defined by an aggregate of >50 cells). Each in vitro assay was performed in duplicate. DNA was obtained from peripheral blood granulocytes from each patient to quantify the JAK2 V617F allele burden at the time of culture assay. Results: LY2784544, at concentrations ranging from 0.03–30.0 μM, inhibited growth of unselected peripheral blood eBFU-E and CFU-GM from PV patients carrying the JAK2 V617F mutation in a dose-dependent manner, although without achieving complete inhibition of all colonies (fig.1). Conclusions: In vitro studies show that LY2784544 decreases the eBFU-E and CFU-GM growth in therapy-naive JAK2 V617F positive PV patients. Our data suggest that LY2784544 may be a candidate for the treatment of MPD carrying the JAK2 V617F mutation. Disclosures: Ma: Eli Lilly and Company: Employment. Walgren:Eli Lilly and Company: Employment.


Blood ◽  
2006 ◽  
Vol 107 (9) ◽  
pp. 3495-3502 ◽  
Author(s):  
Ann Zeuner ◽  
Francesca Pedini ◽  
Michele Signore ◽  
Giusy Ruscio ◽  
Carlo Messina ◽  
...  

Polycythemia vera (PV) is a clonal myeloproliferative disorder characterized by excessive erythrocyte production. Most patients with PV harbor an activating JAK2 mutation, but the molecular links between this mutation and erythrocyte overproduction are unknown. The interaction between death receptors and their ligands contributes to the physiological regulation of erythropoiesis through the inhibition of erythroblast proliferation and differentiation. With the use of an in vitro culture system to generate differentiating erythroid cells, we found that erythroblasts derived from patients with PV harboring the JAK2 V617F mutation were able to proliferate and generate higher numbers of mature erythroid cells in the presence of inhibitory signals delivered by CD95 (Fas/Apo-1) and TRAIL receptor stimulation. JAK2-mutated PV erythroblasts showed lower levels of CD95-induced caspase activation and incomplete caspase-mediated cleavage of the erythroid transcription factor GATA-1, which was entirely degraded in normal erythroblasts on CD95 stimulation. JAK2 mutation was associated in PV erythroblasts with cytokine-independent activation of the JAK2 effectors Akt/PKB and ERK/MAP and with a deregulated expression of c-FLIPshort, a potent cellular inhibitor of death receptor–induced apoptosis. These results show the presence in PV erythroblasts of proliferative and antiapoptotic signals that may link the JAK2 V617F mutation with the inhibition of death receptor signaling, possibly contributing to a deregulation of erythropoiesis.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 262-262
Author(s):  
Hana Bruchova ◽  
Michaela Merkerova ◽  
Eva Otahalova ◽  
Josef T. Prchal

Abstract Polycythemia vera (PV) is a clonal myeloproliferative disorder caused by somatic mutation of a hematopoietic multipotent cell. PV hematopoiesis is characterized by an accumulation of phenotypically normal erythrocytes with overproduction of leukocytes and platelets. A somatic JAK2 V617F point mutation occurs in the majority (>95%) of PV patients. However, this mutation is also found in ∼50% of patients with either essential thrombocythemia (ET) or idiopathic myelofibrosis (MF). The non-specificity of this mutation, the presence of JAK2 V617F-negative PV patients, JAK2 V617F-negative and positive relatives, and evidence of both JAK2 V617F-negative and positive PV clones demonstrate that the JAK2 V617F mutation is not the initial and sole somatic event for the pathogenesis of PV. MicroRNAs (miRNAs) have been shown to be important regulators of hematopoiesis. To identify deregulated miRNAs involved in PV pathogenesis, we studied gene expression of miRNAs of in vitro expanded erythroid progenitors (EPs), peripheral blood mononuclear cells (MNCs), granulocytes, reticulocytes, and platelets from PV patients and healthy controls. Initially, we performed gene expression profiling in 5 PV patients and 5 control cells using CombiMatrix MicroRNA CustomArray with 326 probes. The array data were analyzed by Genesis software to determine differentially expressed miRNAs in PV. These miRNAs were tested in a larger set of samples (n=18) by qRT-PCR and their expression was correlated to the JAK2 V617F mutational level. Hierarchical clustering analysis defined miRNA expression profiles of particular cell lineage for normal and PV cells. Further, ANOVA identified 31 miRNAs differently (P<0.05) expressed in at least some PV lineage cells. Of these miRNAs, we confirmed downregulation of miR-150 in expanded EPs of all stages of maturation, downregulation of let7a and upregulation of miR-182 in PV granulocytes; upregulation of miR-143, miR-145 and miR-223 in PV MNCs; and down-regulation of miR-30b, miR-30c and miR-150 in PV reticulocytes by qRT-PCR. Correlation analysis of miRNA expression with JAK2 V617F mutational level showed a positive correlation of miR-143 (r=0.68) and a negative correlation of let7a (r =−0.63), miR-30c (r=−0.74), miR-342 (r=−0.66) and miR-150 (r=−0.89). To validate PV specificity, we compared expression levels of these miRNAs to other MPD disorders (MF and ET) in which the JAK2 V617F mutation occurs. Putative miRNA targets were predicted by TargetScan 4.0 and PicTar software, and transcript levels of selected target genes are being analyzed to determine their potential deregulation at the mRNA level. Downregulated miR-150 is predicted to the target MYB oncogene that plays an important role in erythropoiesis by maintaining proliferation at the early stages. The most potential target of let7a is HGMA2, whose aberrant expression may contribute to clonal hematopoiesis in PNH. The verification of these predictions is in process by use of miRNA inhibitors, protein levels, and functional studies of the progenitors. Our study demonstrates that the specific signatures of miRNAs define particular peripheral blood cell lineages. Furthermore, the deregulated miRNAs, whose expressions correlate with the JAK2 mutational level, may be associated with the PV phenotype. Understanding the role of deregulated miRNAs in PV should provide insight into the pathogenesis of PV and may lead to novel therapeutic strategies.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1527-1527
Author(s):  
Sai Li ◽  
Robert Kralovics ◽  
Gennaro De Libero ◽  
Andre Tichelli ◽  
Radek C. Skoda

Abstract Mutations in exon 12 of the JAK2 gene have been identified in a minority of patients with myeloproliferative disorders (MPD) and are associated with a selective increase in erythropoiesis resulting in polycythemia vera (PV) or idiopathic erythrocytosis. We compared the lineage distribution of JAK2 mutations in the peripheral blood of 8 PV patients with mutations in exon 12 and 21 PV patients with the JAK2-V617F mutation. Five different exon 12 mutations were observed in the 8 patients studied. Peripheral blood cells were fractionated to obtain granulocytes, platelets and mononuclear cells, which were further separated by FACS into T cells, B cells, NK cells and monocytes. Using a sensitive and quantitative assay to assess the percentages of chromosomes carrying exon 12 mutations, we detected exon 12 mutations in purified granulocytes, monocytes and platelets of all patients studied. A similar distribution was also found for the JAK2-V617F mutation. Exon 12 mutation was absent in sorted lymphoid cells of 5/8 patients, in 2/8 patients only NK cells were positive and in 1/8 patients also B cells carried the mutation. Exon 12 mutations were absent in T cells of all patients studied and JAK2-V617F was present in T cells of only 1/21 patients. Thus, inter-individual differences are notable in the involvement of lymphoid lineages for both exon 12 and JAK2-V617F mutations. To determine the presence of the JAK2 mutations in the erythroid lineage, we performed colony assays in methylcellulose, picked single erythroid colonies grown in the presence or absence of Epo and determined the allelic ratios for each individual colony. In 4/8 patients an exon 12 mutation (E543-D544del) was present in all EECs examined and similarly, in 8/12 patients the JAK2-V617F mutation was found in all EECs. In the remaining patients, we detected some EECs with only the wild type JAK2, suggesting that additional clonal events may also be present in some patients with exon 12 mutations. Interestingly, one patient carried exon 12 and JAK2-V617F mutations. None of the erythroid colonies in this patient carried both mutations simultaneously, indicating that the exon 12 mutation and JAK2-V617F represent two separate clones. One patient displayed erythroid colonies homozygous for the exon 12 mutation and an allelic ratio greater that 50% in granulocytes, indicating that progression to homozygosity can occur in some patients. The lineage distributions of exon 12 mutations and JAK2-V617F are similar and do not explain why exon 12 mutations are associated solely with PV phenotype, whereas JAK2-V617F can also cause essential thrombocythemia or primary myelofibrosis.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2601-2601 ◽  
Author(s):  
Amos S. Gaikwad ◽  
Srdan Verstovsek ◽  
Ko-Tung Chang ◽  
Donghoon Yoon ◽  
William Vainchenker ◽  
...  

Abstract Polycythemia vera (PV) is an acquired myeloproliferative, clonal stem cell disorder characterized by cytokine hypersensitivity. Several groups reported a clonal mutation in the pseudo kinase domain of the Janus kinase 2 (Jak2) protein, substituting phenylalanine at position 617 with valine (V617F) that causes constitutive activation of the JAK/STAT pathway in PV patients. Chronic myelogenous leukemia is another myeloproliferative disorder associated with aberrant tyrosine kinase (bcr-abl) that arises from t(9:22) translocation. Constitutive bcr-abl tyrosine kinase activity suppresses apoptosis leading to survival and proliferation of cells at low cytokine concentrations. Imitanib has been developed to inhibit the bcr-abl kinase activity and has had impressive therapeutic efficacy. However, imitanib also inhibits other tyrosine kinases. Clinical benefits of imitanib in PV have been reported. Mouse FDCP cells over-expressing either the wild-type Jak2 (JAK/W) or two cell lines with the V617F mutation (V617F), one with cytokine hypersensitivity and the other cytokine independent, were created. We examined the effect of imitanib by MTT proliferation assay and propidium iodide staining analysis. No appreciable changes in the proliferation and DNA content were observed in all three cell-lines after imitanib treatment at ~1μM (the concentration effective for the bcr-abl expressing cells and achievable in vivo in imitanib treated patients). However, after 72h of exposure, the cells expressing JAK2 V617F mutant showed 50% inhibition of growth at ~6μM imitanib with no significant effect on the growth of cells expressing JAK/W. To further understand the mechanism of growth inhibition of V617F cells by imitanib, we examined the metabolism of these cells since the constitutively active tyrosine kinases has been demonstrated to change the glucose metabolism. Imitanib treatment (5μM) for 72h caused about 30% decrease in the glucose uptake in V617F cells with only a marginal (~5%) decrease in glucose uptake was observed in the JAK/W cells. We then examined the effect of imitanib on in vitro expanded native human erythroid progenitors (CD71 and CD235a positive cells) from three normal and four PV individuals who expressed variable proportion of mutant JAK2 V617F alleles. In this small study, between 18–30% decrease in the proliferation of the cells from PV patients was seen with 1–2μM of imitanib compared to 8% seen with normal erythroid progenitors; however, the in vitro expansion was accompanied by a decrease of proportion of cells with JAK2 mutation (see abstract Prchal et. Al. at this meeting). Further, in ongoing clinical studies, one of 5 tested PV patients who showed an excellent clinical response to imitanib therapy had decrease of V617F mutation from 58 to 19% in circulating granulocytes. We conclude that high concentration of imitanib is required to achieve the cytotoxic effects in reporter cells transfected with JAK2 mutation that are not readily achievable in vivo; however, lower doses (but still significantly higher than those needed for bcr-abl expressing cells) are effective in native PV progenitors. These data do not exclude possible useful therapeutic effect of imitanib in PV either alone or in combination with other drugs. Amos Gaikwad, Ph.D. and Srdan Verstovsek, M.D. contributed equally to this study.


Cell Research ◽  
2008 ◽  
Vol 18 (S1) ◽  
pp. S141-S141
Author(s):  
Alvin CH Ma ◽  
Alice MS Cheung ◽  
Alister C Ward ◽  
Wing-Yan Au ◽  
Yok-Lam Kwong ◽  
...  

Blood ◽  
2006 ◽  
Vol 107 (9) ◽  
pp. 3676-3682 ◽  
Author(s):  
Francesco Passamonti ◽  
Elisa Rumi ◽  
Daniela Pietra ◽  
Matteo G. Della Porta ◽  
Emanuela Boveri ◽  
...  

We studied the relationship between granulocyte JAK2 (V617F) mutation status, circulating CD34+ cells, and granulocyte activation in myeloproliferative disorders. Quantitative allele-specific polymerase chain reaction (PCR) showed significant differences between various disorders with respect to either the proportion of positive patients (53%-100%) or that of mutant alleles, which overall ranged from 1% to 100%. In polycythemia vera, JAK2 (V617F) was detected in 23 of 25 subjects at diagnosis and in 16 of 16 patients whose disease had evolved into myelofibrosis; median percentages of mutant alleles in these subgroups were significantly different (32% versus 95%, P < .001). Circulating CD34+ cell counts were variably elevated and associated with disease category and JAK2 (V617F) mutation status. Most patients had granulocyte activation patterns similar to those induced by administration of granulocyte colony-stimulating factor. A JAK2 (V617F) gene dosage effect on both CD34+ cell counts and granulocyte activation was clearly demonstrated in polycythemia vera, where abnormal patterns were mainly found in patients carrying more than 50% mutant alleles. These observations suggest that JAK2 (V617F) may constitutively activate granulocytes and by this means mobilize CD34+ cells. This exemplifies a novel paradigm in which a somatic gain-of-function mutation is initially responsible for clonal expansion of hematopoietic cells and later for their abnormal trafficking via an activated cell progeny.


2005 ◽  
Vol 280 (51) ◽  
pp. 41893-41899 ◽  
Author(s):  
Judith Staerk ◽  
Anders Kallin ◽  
Jean-Baptiste Demoulin ◽  
William Vainchenker ◽  
Stefan N. Constantinescu

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2578-2578
Author(s):  
Daniela Pietra ◽  
Alessandra Balduini ◽  
Carmela Marseglia ◽  
Matteo G. Della Porta ◽  
Luca Malcovati ◽  
...  

Abstract A unique gain-of-function mutation of the Janus kinase 2 (JAK2) gene has been recently described in patients with polycythemia vera (PV), essential thrombocythemia and chronic idiopathic myelofibrosis [N Engl J Med. 2005 Apr 28;352(17):1779–90]. Although the currently available data clearly demonstrate that the JAK2 V617F mutation participates in the pathogenesis of myeloproliferative disorders, the mutation’s precise place in the hierarchical order of pathogenetic events remains to be established. We have recently reported that altered gene expression in myeloproliferative disorders correlates with activation of signaling by the V617F mutation of JAK2 (Blood. 2005 Aug 4; Epub ahead of print). Granulocyte CD177 (PRV1) mRNA overexpression has been initially reported as a potential marker of PV but later shown by us to rather be a marker of neutrophil activation [Br J Haematol. 2004 Sep;126(5):650–6]. In this study, we analyzed the relationship between JAK2 V617F mutation status, granulocyte CD177 mRNA expression and CD177 soluble protein level in 72 patients with PV. We also investigated the ontogeny of CD177 expression by hematopoietic cells with the aim of defining the stage of mRNA expression during myeloid, erythroid and megakaryocytic cell differentiation. Finally we studied the effect of soluble CD177 protein on hematopoietic cell proliferation and differentiation. Granulocyte CD177 mRNA expression and percentage of JAK2 V617F alleles were evaluated by quantitative Real Time PCR (qRT-PCR), while serum CD177 protein level was measured by a flow cytometry-based competitive antibody-binding assay. Liquid cultures were performed by culturing peripheral blood mononuclear cells obtained from healthy individuals and PV patients in the presence of high CD177-expressing, low CD177-expressing or CD177-depleted sera. After 12 days of culture, cells were collected, counted and evaluated for colony growth, and for flow cytometry analysis of myeloid, erythroid, megakaryocytic and CD34-positive cell subpopulations. qRT-PCR studies showed a close relationship between CD177 mRNA level and percentage of JAK2 V617F alleles (r=0.412, P&lt;0.001). CD177 mRNA expression was almost undetectable in cell populations other than granulocytes. Studies of CFU-GM growth and differentiation indicated that CD177 mRNA expression is a late event restricted to the neutrophil stage of differentiation. Analysis of serum samples showed variable values for mean fluorescence intensity (MFI), indicating variable levels of the soluble CD177 protein in the patients studied. A very close relationship was found between granulocyte CD177 mRNA expression and soluble CD177 protein level (r=0.56, P=0.02). Incubation of mononuclear cells with serum samples showing high levels of soluble CD177 protein resulted in increased numbers of CD34-positive cells (P&lt;0.02) and of erythroid progenitors (P&lt;0.03). This effect was not detectable when low CD177-expressing or CD177-depleted sera were employed. These observations clearly indicate that the JAK2 V617F mutation is associated with enhanced granulocyte CD177 mRNA expression, and that this latter results in high levels of soluble CD177 protein. These elevated levels might contribute to the increased red cell production that characterizes polycythemia vera.


Sign in / Sign up

Export Citation Format

Share Document