Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 485-485
    Abstract: The pathologic hallmark of systemic mastocytosis (SM) is differentiation and cluster formation of mast cells (MC) in hematopoietic tissues. The somatic c-kit mutation D816V is detectable in a majority of all SM patients independent of the proliferation-status of MC or subtype (indolent or aggressive) of disease. To investigate the role of c-kit D816V in the pathogenesis of SM, we established a Ba/F3 cell line with doxycycline-inducible expression of c-kit D816V. We found that c-kit D816V provides a strong signal for mast cell differentiation and cluster formation in Ba/F3 hematopoietic progenitor cells without enhancing their growth thereby resembling the clinical presentation of indolent SM (ISM). As assessed by gene chip analysis, induction of c-kit D816V resulted in expression of various differentiation antigens including mouse mast cell protease 5, mi transcription factor, histidine decarboxylase (HDC), secretory granule proteoglycan, IL-4 receptor, ICAM-1, and CD63 consistent with an early phase of mastopoiesis. By contrast, c-kit D816V did neither induce expression of granulo-monocytic antigens such as myeloperoxidase, IL-3 receptor, or GM-CSF receptor, nor expression of ‘late stage’ mast cell antigens such as FcεRI. The c-kit D816V-induced synthesis of histamine in Ba/F3 cells was confirmed by RIA. To examine the role of c-kit D816V in the pathogenesis of mastocytosis, we extended our analysis to bone marrow biopsy sections obtained from patients with ISM. In these experiments, the D816V-mutated form of c-kit was detected more frequently in micro-dissected tryptase-positive MC obtained from dense compact infiltrates (44.2%) than in diffusely spread MC in these patients (22.6%) (p & lt;0.05). In summary, our data establish a role for c-kit D816V in differentiation and cluster formation of neoplastic (mast) cells. Additional genetic hits, apart from c-kit D816V, may be responsible for aggressive growth of MC in advanced MC neoplasms.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: The American Journal of Surgical Pathology, Ovid Technologies (Wolters Kluwer Health), Vol. 28, No. 10 ( 2004-10), p. 1319-1325
    Type of Medium: Online Resource
    ISSN: 0147-5185
    RVK:
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2004
    detail.hit.zdb_id: 2029143-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 2917-2917
    Abstract: Abstract 2917 Poster Board II-893 BACKGROUND: Myeloproliferative neoplasms (MPN) are hematopoietic stem cell diseases encompassing chronic myeloid leukemia (CML), essential thrombocythemia (ET), idiopathic myelofibrosis (IMF), and polycythemia vera (PV). More than 95% of all patients with PV and some 50% of patients with ET and IMF have an activating mutation of JAK2 (JAK2 V617F). Tumor necrosis factor-alpha (TNF) is a pleiotropic cytokine produced by multiple cell types, including macrophages, NK, T and B cells. TNF plays a key regulatory role in immune and inflammatory responses. There is also evidence that TNF is involved in the regulation of erythropoiesis. We have previously reported that TNF levels are elevated in mice with retrovirally induced JAK2 V617F-positive PV-like MPN and that lack of TNF attenuates MPN in this model. APPROACH AND RESULTS: We studied the expression of TNF in white blood cells from PV patients (n=43) and normal controls (n=21), using qPCR. PV patients expressed on average 2.5-fold higher levels of TNF than controls (p 〈 0.0004). Upon examination of a panel of human leukemia lines (n=8) we found the highest TNF expression in HEL cells, which are homozygous for JAK2 V617F, suggesting that JAK2 V617F may upregulate TNF expression. Consistent with this, we found 6.9-fold higher TNF expression in Ba/F3 cells constitutively co-expressing EPOR and JAK2 V617F compared to parental cells. Exposure of HEL cells or Ba/F3 EPOR JAK2 V617F cells to the JAK2 inhibitor CYT387 (2 μM) resulted in a time dependent 3.3 to 7.5-fold decrease of TNF mRNA as assessed by qPCR. By contrast, TNF mRNA expression was not found to be down-regulated by CYT387 in HL60 cells lacking mutated JAK2. CYT387 was also found to down-regulate TNF mRNA expression in MNC derived from MPN patients. To assess differential effects of TNF on normal versus MPN progenitor cells, we performed clonogenic assays of peripheral blood MNC from normal donors (n=4) and MNC from MPN patients (n=9) in the presence of graded concentrations of EPO (0, 0.05, 0.5 and 5 IU/mL) and TNF (1, 10, 100 ng/mL). Intermediate and high TNF (10, 100 ng/mL) caused a dose-dependent reduction of BFU-E and CFU-GM compared to controls. However, in all conditions colony survival in MPN samples was higher compared to normal controls. Low TNF (1 ng/mL) in cultures supported by EPO (0.5 or 5.0 IU/mL) increased BFU-E formation by MPN cells to 142% of controls, but reduced BFU-E formation by normal MNC to 65%. CONCLUSIONS: Our data indicate that JAK2 V617F upregulates TNF expression in cell lines and primary MPN cells. Compared to normal progenitor cells, MPN progenitor cells are less sensitive to or even stimulated by TNF. These data suggest that JAK2 V617F-induced TNF may contribute to MPN pathogenesis by conferring a growth advantage to MPN over normal cells. Disclosures: Druker: OHSU patent #843 - Mutate ABL Kinase Domains: Patents & Royalties; MolecularMD: Equity Ownership; Roche: Consultancy; Cylene Pharmaceuticals: Consultancy; Calistoga Pharmaceuticals: Consultancy; Avalon Pharmaceuticals: Consultancy; Ambit Biosciences: Consultancy; Millipore via Dana-Farber Cancer Institute: Patents & Royalties; Novartis, ARIAD, Bristol-Myers Squibb: Research Funding. Burns:Cytopia: Employment. Deininger:Genzyme: Research Funding; BMS: Consultancy; Novartis: Consultancy, Honoraria; Ariad : Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 1987-1987
    Abstract: Chronic myeloid leukemia (CML) is a myeloproliferative disease in which BCR/ABL enhances survival of leukemic cells through modulation of pro- and anti-apoptotic molecules. Recent data suggest that pro-apoptotic Bim plays a role as a tumor-suppressor in myeloid cells, and that leukemic cells express only low amounts of this death activator. In the current study, we have investigated expression of Bim in primary CML cells and in the CML cell lines K562 and KU812, and in Ba/F3 cells inducibly expressing BCR/ABL on exposure to doxycycline (TonB.210-X). As assessed by Northern- and Western blotting, primary CML cells were found to express significantly lower amounts of Bim mRNA and Bim protein compared to normal bone marrow cells. The BCR/ABL-inhibitors imatinib (Novartis Pharma AG) and AMN107 (Novartis Pharma AG) were found to promote Bim expression in CML cells at pharmacologic concentrations. Correspondingly, BCR/ABL was found to down-regulate expression of Bim in TonB.210-X cells. The BCR/ABL-induced decrease in expression of Bim in leukemic cells was found to be a post-transcriptional event that depended on signaling through MEK, and was abrogated by the proteasome-inhibitor MG132. Interestingly, MG132 was found to up-regulate Bim-expression and to suppress the growth of Ba/F3 cells containing either wild-type BCR/ABL or various imatinib-resistant mutants of BCR/ABL including the T315I mutant that is resistant to all currently available ATP-competitive tyrosine kinase inhibitors (IC50: 30–100 nM). To confirm the role of Bim as a tumor suppressor in CML, a Bim specific siRNA was transfected into K562 cells. This siRNA was found to counteract imatinib- and MG132-induced cell death. In conclusion, our data identify BCR/ABL as a Bim-suppressor in CML cells and suggest, that re-expression of Bim by proteasome inhibition or by targeting of signaling pathways downstream of BCR/ABL may be an attractive therapeutic approach in imatinib-resistant CML.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 1986-1986
    Abstract: Chronic myeloid leukemia (CML) is a myeloproliferative disease characterized by the BCR/ABL oncogene and an increased survival of leukemic cells. The BCR-ABL tyrosine kinase inhibitor imatinib has successfully been introduced as a treatment of CML. However, resistance after an intitial response is common in patients with advanced disease, and it is not yet clear if responses in early disease phases will be durable. Therefore, current studies focus on novel potential drug-targets in CML cells. We have recently identified heme oxygenase-1 (HO-1) as a novel BCR/ABL-dependent survival-molecule in primary CML cells. In this study, we analyzed signal transduction pathways underlying BCR/ABL-induced expression of HO-1 and evaluated the role of HO-1 as a potential new target of drug therapy. We found that the PI3-kinase inhibitor LY294002 and MEK inhibitor PD98059 downregulate expression of HO-1 in CML cells. In addition, constitutively active Ras- and Akt -mutants were found to promote expression of HO-1 in Ba/F3 cells, further supporting the involvement of the PI3-kinase/Akt as well as the MAPK pathway in regulating HO-1 expression. To establish a role for HO-1 in survival of CML cells, expression of HO-1 was silenced by siRNAs which resulted in apoptosis of K562 cells. Next, HO-1 was targeted in CML cells by pegylated zinc protoporphyrin (PEG-ZnPP), a competitive inhibitor of HO-1. Exposure to PEG-ZnPP resulted in growth inhibition and induction of apoptosis in primary CML cells as well as in the CML-derived cell lines K562 and KU812 with IC50 values ranging between 1–10 μM. The growth-inhibitory effects of PEG-ZnPP were not only observed in CML cells responsive to imatinib, but also in imatinib-resistant K562 cells and Ba/F3 cells expressing various imatinib-resistant mutants of BCR/ABL (T315I, E255K, M351T, Y253F, Q252H, H396P). Moreover, imatinib and PEG-ZnPP were found to exert synergistic growth inhibitory effects on imatinib-resistant leukemic cells. Together, these data suggest that HO-1 represents a novel drug target in cells expressing BCR/ABL, including those with resistance to imatinib.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 118, No. 24 ( 2011-12-08), p. 6392-6398
    Abstract: Proinflammatory cytokines such as TNFα are elevated in patients with myeloproliferative neoplasms (MPN), but their contribution to disease pathogenesis is unknown. Here we reveal a central role for TNFα in promoting clonal dominance of JAK2V617F expressing cells in MPN. We show that JAK2V617F kinase regulates TNFα expression in cell lines and primary MPN cells and TNFα expression is correlated with JAK2V617F allele burden. In clonogenic assays, normal controls show reduced colony formation in the presence of TNFα while colony formation by JAK2V617F-positive progenitor cells is resistant or stimulated by exposure to TNFα. Ectopic JAK2V617F expression confers TNFα resistance to normal murine progenitor cells and overcomes inherent TNFα hypersensitivity of Fanconi anemia complementation group C deficient progenitors. Lastly, absence of TNFα limits clonal expansion and attenuates disease in a murine model of JAK2V617F-positive MPN. Altogether our data are consistent with a model where JAK2V617F promotes clonal selection by conferring TNFα resistance to a preneoplastic TNFα sensitive cell, while simultaneously generating a TNFα-rich environment. Mutations that confer resistance to environmental stem cell stressors are a recognized mechanism of clonal selection and leukemogenesis in bone marrow failure syndromes and our data suggest that this mechanism is also critical to clonal selection in MPN.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 65, No. 20 ( 2005-10-15), p. 9436-9444
    Abstract: Chronic myeloid leukemia (CML) is a myeloproliferative disease in which BCR/ABL enhances survival of leukemic cells through modulation of proapoptotic and antiapoptotic molecules. Recent data suggest that proapoptotic Bcl-2–interacting mediator (Bim) plays a role as a tumor suppressor in myeloid cells, and that leukemic cells express only low amounts of this cell death activator. We here show that primary CML cells express significantly lower amounts of bim mRNA and Bim protein compared with normal cells. The BCR/ABL inhibitors imatinib and AMN107 were found to promote expression of Bim in CML cells. To provide direct evidence for the role of BCR/ABL in Bim modulation, we employed Ba/F3 cells with doxycycline-inducible expression of BCR/ABL and found that BCR/ABL decreases expression of bim mRNA and Bim protein in these cells. The BCR/ABL-induced decrease in expression of Bim was found to be a posttranscriptional event that depended on signaling through the mitogen-activated protein kinase pathway and was abrogated by the proteasome inhibitor MG132. Interestingly, MG132 up-regulated the expression of bim mRNA and Bim protein and suppressed the growth of Ba/F3 cells containing wild-type BCR/ABL or imatinib-resistant mutants of BCR/ABL. To show functional significance of “Bim reexpression,” a Bim-specific small interfering RNA was applied and found to rescue BCR/ABL-transformed leukemic cells from imatinib-induced cell death. In summary, our data identify BCR/ABL as a Bim suppressor in CML cells and suggest that reexpression of Bim by novel tyrosine kinase inhibitors, proteasome inhibition, or by targeting signaling pathways downstream of BCR/ABL may be an attractive therapeutic approach in imatinib-resistant CML.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2005
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 115, No. 25 ( 2010-06-24), p. 5232-5240
    Abstract: Activating alleles of Janus kinase 2 (JAK2) such as JAK2V617F are central to the pathogenesis of myeloproliferative neoplasms (MPN), suggesting that small molecule inhibitors targeting JAK2 may be therapeutically useful. We have identified an aminopyrimidine derivative (CYT387), which inhibits JAK1, JAK2, and tyrosine kinase 2 (TYK2) at low nanomolar concentrations, with few additional targets. Between 0.5 and 1.5μM CYT387 caused growth suppression and apoptosis in JAK2-dependent hematopoietic cell lines, while nonhematopoietic cell lines were unaffected. In a murine MPN model, CYT387 normalized white cell counts, hematocrit, spleen size, and restored physiologic levels of inflammatory cytokines. Despite the hematologic responses and reduction of the JAK2V617F allele burden, JAK2V617F cells persisted and MPN recurred upon cessation of treatment, suggesting that JAK2 inhibitors may be unable to eliminate JAK2V617F cells, consistent with preliminary results from clinical trials of JAK2 inhibitors in myelofibrosis. While the clinical benefit of JAK2 inhibitors may be substantial, not the least due to reduction of inflammatory cytokines and symptomatic improvement, our data add to increasing evidence that kinase inhibitor monotherapy of malignant disease is not curative, suggesting a need for drug combinations to optimally target the malignant cells.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: The FASEB Journal, Wiley, Vol. 19, No. 8 ( 2005-06), p. 960-962
    Type of Medium: Online Resource
    ISSN: 0892-6638 , 1530-6860
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2005
    detail.hit.zdb_id: 1468876-1
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 795-795
    Abstract: Abstract 795 Elevated levels of pro-inflammatory cytokines such as TNF-alpha (TNF) have been implicated in the constitutional symptoms commonly seen in JAK2 V617F MPN. However, it is unclear to what extent they contribute to the disease process itself. We hypothesized that JAK2 V617F may confer a selective advantage on hematopoietic progenitors residing in the high cytokine milieu uniquely present MPN patients, focusing on the pro-inflammatory cytokine TNF. We used ELISA and qPCR to measure TNF in plasma and neutrophils of MPN patients and normal controls. All MPN subtypes had increased plasma concentrations of TNF, with primary myelofibrosis, polycythemia vera and essential thrombocythemia displaying 6, 4 and 2 fold higher mean concentrations of plasma TNF as compared to normal controls, respectively. Median neutrophil TNF mRNA was 2 fold higher in PV patients as compared to normal controls (p 〈 0.001). We then tested the effect of TNF on CFU-GM and BFU-E formation from MPN vs normal mononuclear cells. At low concentrations (1ng/ml) of TNF we observed a decrease in BFU-E from normal controls to 82% of that without TNF but an increase in MPN BFU-E to 120% (p 〈 0.05) of that without TNF. We also observed a blunting of TNF induced CFU-GM suppression in MPN versus normal. We genotyped colonies and found that in the presence of TNF colony formation was almost exclusively JAK2 V617F, demonstrating that TNF selects for JAK2 V617F colony growth. To distinguish between direct and indirect TNF effects (i.e. effects mediated through bystander cells) as well as to potentially augment the differential effects of TNF on normal and MPN by the use of a more homogenous cell population we performed equivalent colony formation experiments on CD34+ cells. The most notable difference observed was the effect 10ng/ml TNF on CFU-GM. TNF reduced CFU-GM colony formation by normal CD34+ cells to 50%, but increased CFU-GM colony formation by MPN cells to 120% (p 〈 0.001). The suppressive effect of 10ng/ml on BFU-E was also blunted in MPN versus normal CD34+ cells (49% vs 30% of that without TNF) (p=0.054). Genotyping of MPN colonies allowed us to compare the effect of TNF on colony formation on three distinct populations: (1) JAK2WT MPN CD34+ cells, (2) JAK2V617F MPN CD34+ cells and (3) normal CD34+ cells. As compared to normal controls (59% decrease), JAK2 non-mutated MPN CD34+ cells are hypersensitive to myeloid colony suppression (78% decrease) whereas TNF paradoxically enhances colony formation by JAK2 V617F CD34+ cells (180% increase). This suggests that “pre-JAK2” CD34+ cells are functionally disadvantaged when compared to those from normal controls and implicates TNF in the selection and clonal expansion of the JAK2 V617F clone. To confirm the latter we tested whether ectopic expression of JAK2 V617F in murine hematopoietic progenitors blunts TNF-induced inhibition of myeloid colony formation. In the presence of TNF, colony formation of hematopoietic progenitors expressing empty vector or BCR-ABL (included as an additional control) was decreased to 60%, while TNF had no effect on progenitors with ectopic expression of JAK2 V617F. This suggests that JAK2 V617F directly imparts TNF resistance. To interrogate the role of TNF for the development of MPN in vivo, we infected TNF+/+ and TNF−/− bone marrow with JAK2 V617F-GFP retrovirus and transplanted the cells into genotypically matched recipients. Two strains of mice were used in independent experiments (C57BL6 and B6129S6). Equal numbers of TNF+/+ and TNF−/− GFP+ cells per mouse were injected. Although peripheral blood counts were comparable, JAK2 V617F burden (assessed by GFP%) was significantly lower in recipients of TNF−/− knockout bone marrow compared to recipients of TNF+/+ marrow. This difference in JAK2 V617F burden between the two groups was apparent in peripheral blood (47% vs 3.7%, p 〈 0.05) and bone marrow (52% vs 2%, p 〈 0.05) and the spleens of the TNF−/− were smaller (0.46g vs 0.11g, p=0.078). Our data suggest that JAK2 V617F-induced TNF resistance may drive the expansion of the mutant clone by conferring a growth advantage to JAK2 V617F cells over their JAK2 wild-type counterparts in the high cytokine microenvironment of MPN patients. Further, JAK2WT MPN cells exhibit intrinsic TNF hypersensitivity compared to normal cells, which greatly enhances this selective advantage. This JAK2V617F-negative state may play a role in the predisposition to acquire MPN. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages