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. 124, No. 21 ( 2014-12-06), p. 1876-1876
    Abstract: Introduction: Deregulated signal transduction pathways driven by the constitutively active JAK2-V617F kinase, play a central role in pathogenesis of the classical Philadelphia negative myeloproliferative neoplasms (MPNs). Polycythemia vera, essential thrombocythemia and primary myelofibrosis are characterized by clonal proliferation of myeloid cells and development of inflammatory syndrome (CRP elevation, fever, high proinflammatory cytokine plasma levels) particularly, in advanced phases of the disease. Integrins are heterodimeric cell surface receptors that mediate a wide range of anchorage dependent events fundamentally essential in adhesion and mobility of leukocytes during inflammatory states. An intricate network of intracellular signaling pathways triggers the activation of intergins by allosteric conformational changes and/or by altering lateral mobility to increase receptor clustering. Methods: We investigated the role of JAK2-V617F in the regulation of β1 integrin activation, function and expression in myeloid cells using in vitro cell culture models, in vivo JAK2-V617F knock-in model, murine bone marrow transplantation model and finally, clinical patient samples (collected after informed consent obtained in accordance with the Declaration of Helsinki). Integrin activation was studied using flow cytomery based soluble ligand binding assay and by analyzing the expression of 9EG7 activation epitope. The functional static adhesion assay was performed on ligand coated plates and data was acquired using a fluorescence microplate reader. Surface expression of integrins was investigated by flow cytometry. Results: To identify a potential biological impact of JAK2-V617F on β1 integrins, we generated a 32D murine myeloid progenitor cell line transfected with EpoR and either wildtype JAK2 or JAK2-V617F. Under serum starved conditions, the presence of the oncogenic JAK2-V617F mutation strongly induced activation of β1 integrins as revealed by a 6-fold rise in binding to its soluble ligand, VCAM-1-Fc and enhanced exposure of the 9EG7 high-affinity conformation-specific epitope. This also translated into increased adhesion on VCAM-1 in comparison to wildtype cells. Importantly, inhibition of the constitutive JAK2-V617F kinase activity by JAK inhibitor treatment, significantly downregulated β1 integrin activation as well as cellular adhesion. Furthermore, shRNA based knockdown of the endogenous JAK2-V617F expression in HEL (Human Erythroleukemia) cells, reduced adhesion on VCAM-1, thereby underscoring the functional role of the JAK2-V617F kinase in the regulation of β1 integrins. Interestingly, the cell surface expression of β1 integrins remains unaltered in the presence of JAK2-V617F or inhibitor treatment in the 32D cells as well as, following genetic inhibition in HEL cells. Additionally, in an in vivo retroviral bone marrow transplantation model in Balb/c mice, the overexpression of wildtype or JAK2-V617F, showed similar β1 integrin expression in granulocytes. Currently, in vivo studies in a JAK2-V617F knock-in murine model are under way. We then extended our study to investigate the role of constitutively active JAK2-V617F in integrin function in MPN patients. Again, no significant difference was found in β1 integrin expression on primary granulocytes from JAK2-V617F positive MPN patients and age-matched healthy donors. However, granulocytes from JAK2-V617F-positive patients showed higher activation of β1 integrins as exhibited by a 6-fold increase in binding to soluble VCAM-1-Fc when compared to healthy donors. In addition, preliminary analysis suggests stronger adhesion of JAK2-V617F positive granulocytes on VCAM-1. In an attempt to dissect the underlying molecular mechanism, we uncovered a previously undescribed role of JAK2 in the inside-out signaling of integrin activation in 32D myeloid cells. Overexpression of wildype JAK2 or V617F resulted in activation of the small GTPase Rap1, a key integrin regulator, and was subsequently downregulated upon treatment with JAK inhibitor. Conclusions: Taken together, our study highlights a unique and important role of JAK2-V617F in regulation of β1 integrin activation in myeloid cells. This may have functional consequences in pathophysiology of the disease, particularly, in dysregulation of leukocyte adhesion during thrombosis and in induction of inflammation. 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: 2014
    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: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 1045-1045
    Abstract: Abstract 1045 In patients with FLT3-ITD mutated AML, FLT3-inhibitors have been used successfully as a ‘bridging therapy’ before allogeneic transplantation. Inhibitors of other kinases (such as imatinib for BCR-ABL positive CML) have previously been used successfully after allogeneic transplantation – even before discontinuation of immunosuppressive medication. However, it is known that some BCR-ABL inhibitors such as dasatinib exert strong inhibitory effects on primary T-cells through inhibition of Src-kinases relevant for T-cell receptor signaling. Even imatinib and nilotinib - although not affecting Src kinase activity – showed decreased T-cell activation and reactivity to some extent. Thus, the influence of FLT3-kinase inhibitors on T-cell function may be critical in the context of allogeneic bone marrow transplantation for FLT3-ITD-positive AML. Besides inhibition of FLT3-kinase, midostaurin (PKC412) exerts activity against PDGFR, VEGFR or c-KIT. In contrast, second generation inhibitors such as quizartinib (AC220) act in a far more FLT3-specific manner. Therefore, we aimed to investigate the effects of both clinically relevant FLT3-inhibitors on T-cell receptor signaling in comparison to the well characterized and potent BCR-ABL inhibitor dasatinib. Investigating primary T-cells derived from healthy donors, we applied a dose range of 10–50 nM dasatinib, 5–50nM midostaurin and 10–50 nM quizartinib. These dose ranges have been previously described to be achievable as trough levels during inhibitor therapy in early clinical trials. Upon incubation with dasatinib (10nM and 50nM), we found overall reduction in global tyrosine phosphorylation as detected by Western-blotting using the 4G10 antibody. In contrast, treatment with midostaurin left the activation of T-cell receptor signaling pathways unaffected. Comparable to DMSO control, overall phosphorylation was induced almost immediately after stimulation. Western-blotting of LCK and Plcg1 showed similar time dependent activation compared to total phosphorylation. Likewise, quizartinib did not reduce overall tyrosine phosphorylation level and left activation of downstream kinases (ZAP70, MAPK, LCK, Plcg1) largely unaffected. As activation of primary T-cells is a critical step in immune responses against viral and tumor antigens we aimed to investigate the influence of FLT3-kinase inhibitors quizartinib and midostaurin on activation of CD8+ T-cells. T-cells from healthy donors were stimulated using either PHA 0.5% or CD3/CD28 beads to ensure a more T-cell receptor specific stimulation. Using CD3/CD28 stimulation, CD69 expression was almost abrogated following dasatinib treatment. Applying clinically relevant doses of midostaurin or quizartinib to isolated T-cells did not influence CD69 expression. Expression levels upon PHA or CD3/CD28 stimulation were comparable to DMSO-control - even in the presence of 50nM midostaurin or quizartinib. Proliferation of T-cells upon CD3/CD28 stimulation was impaired by dasatinib treatment, while midostaurin and quizartinib left T-cell proliferation largely unaffected – as determined by CSFE staining. In order to investigate the T cell allo-reactivity, mixed lymphocyte culture was performed, where human pan-T-cells are co-cultured with allogeneic antigen presenting cells. T-cell proliferation – as measured by 3H-thymidine incorporation – was significantly impaired by dasatanib but neither midostaurin nor quizartinib treatment. Investigation of leukemia- and virus-antigen-specific T-cell responses are currently under way to gain deeper insight regarding this clinically relevant scenario. Overall, we found FLT3-kinase inhibitors midostaurin and quizartinib to leave T-cell activation, proliferation and function unaffected in-vitro. This information may be useful for the design of up-coming clinical trials testing the safety and efficacy of FLT3-kinase inhibitors in combination with allogeneic stem-cell transplantation. Disclosures: Lipka: Novartis Inc.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Heidel:Novartis Inc.: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 1202-1202
    Abstract: Several genes and signaling pathways control the fine balance between self-renewal and differentiation in hematopoietic stem cells and potentially also in leukemic stem cells (LSC). Phospholipase C family members are key mediators of calcium signaling which play an important role in differentiation and proliferation of immune cells but also contribute to malignant transformation and tumorigenesis. Plcg1 is highly expressed in hematopoietic stem- and progenitor cells and also in myeloid leukemia. Plcg1 gets activated by cell extrinsic receptor stimulation and integrates signals from the cell surface. Its influence on proliferation and differentiation of hematopoietic cells may be largely independent of other bone fide mediators of self-renewal and stem cell viability such as STAT-, MEK-ERK or AKT-signaling. To which extent Plcg1-dependent signal integration is required for function and maintenance of leukemic stem cells remained so far elusive. Genetic inactivation of Plcg1 by RNAi in human AML cell lines led to decreased proliferative capacity. Likewise, knockdown of Plcg1 in AML1-ETO (AML1ETO9a) transformed murine LSK-cells resulted in reduced colony formation and decreased re-plating capacity. In order to validate these findings and to investigate the impact of Plcg1 on myeloid leukemia stem cell function, we generated a conditional knockout mouse model for Plcg1 with Exons 3-5 being flanked with loxP sites. Excision of the respective sequence by activation of a Cre-recombinase resulted in complete loss of a functional protein and transcript. LSK-cells from Plcg1f/f and Plcg1+/+ littermate controls were retrovirally infected with two different oncogenes: either MLL-AF9 (MA9-GFP) or AML1-ETO9a in combination with KRAS (AE9a/KRAS-GFP). Primary recipient mice were injected with GFP+ LSK-cells and monitored for disease development. GFP+ Kit+ cells were isolated from leukemic mice and transduced with a Cre-recombinase, followed by plating in methylcellulose. Inactivation of Plcg1 in AE9a/KRAS transformed cells significantly reduced the number of colonies and decreased re-plating capacity to three rounds. Loss of Pclg1 in MA9 transformed LSC resulted in decreased colony numbers and colony size, however, re-plating capacity was not affected to a major extent. To assess for the requirement of Plcg1 in maintenance of fully developed leukemia, we injected equal numbers of GFP+ Kit+ cells (Plcg1-/- or Plcg1+/+) into sublethally irradiated secondary recipients. Inactivation of Plcg1 was highly deleterious for AE9a/KRAS induced AML-LSC and reduced disease penetrance by more than 85%. Depletion of Plcg1 in MA9 transformed cells delayed AML development and significantly prolonged survival of recipient mice. Leukemias that developed from Plcg1-/- donors showed complete excision of Plcg1, indicating, that Plcg1 deficient leukemia can develop in an MLL-AF9 driven background. However, when transplanting MA9 transformed Plcg1-/- or Plcg1+/+ bone marrow cells into tertiary recipient mice, loss of Plcg1 significantly delayed disease progression and reduced disease penetrance. To quantify this loss of leukemic stem cells, we performed limiting dilution analysis using purified LSCs from diseased Plcg1-/- or Plcg1+/+ MA9 secondary recipient mice. LSC frequency was markedly reduced in tertiary recipients of Plcg1-depleted LSCs (1 in 78,000 Plcg1-/- vs. 1 in 3,000 Plcg1+/+). Genetic inactivation of Plcg1 in LSCs derived from primary recipient mice (either MA9 or AE9a/KRAS driven AML) led to induction of differentiation as assessed by cell morphology and immunophenotyping, and this effect was more pronounced in AE9a/KRAS transformed cells. To investigate whether transcriptional effectors of Plcg1 signaling affect the fine balance between self-renewal in MA9- and AE9a/KRAS-driven leukemia, we performed whole transcriptome analysis (RNAseq) on sorted LSCs. Ongoing analyses address the functional difference between AML-ETO and MLL-AF9 driven disease and elucidate on distinct patterns of activated gene sets depending on the oncogenic background. Taken together, Plcg1 is required for maintenance of myeloid leukemia stem cells. Understanding of its relevance in LSC biology and function may offer the opportunity to develop this relevant signaling node as a target structure in AML. 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: 2016
    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. 134, No. Supplement_1 ( 2019-11-13), p. 529-529
    Abstract: Several cellular pathways control the fine balance between self-renewal and differentiation to maintain leukemia-initiating cell (LIC) function. To identify cellular dependencies with relevance for oncogenic fusion proteins, we performed global proteome profiling. Acute myeloid leukemia (AML) was induced by retroviral expression of either MLL-AF9 (MA9) or AML1-ETO9a (AE) in murine hematopoietic stem and progenitor cells (HSPCs) (Lineage-Sca1+Kit+, LSK) which were subsequently transplanted into irradiated syngeneic recipients. After onset of leukemia, LIC-enriched (GFP+ Kithigh) cells isolated from 4 different primary recipients (per oncogene) were analyzed by in-depth quantitative proteomic analysis using high-resolution mass spectrometry (MS). More than 3,000 proteins were quantified with 868 proteins being differentially expressed between MA9 and AE LIC-enriched populations. In MLL-rearranged (MLLr) cells, gene set enrichment analysis (GSEA) revealed significant enrichment of cellular functions related to protein degradation and proteasome function. As this enrichment is present in MLLr-leukemia but not AE-driven LICs, may indicate an oncogene specific vulnerability. Expression of proteasome subunits is highly heterogeneous between different cell types and therefore may also be influenced by the underlying differentiation stage or oncogenic fusion. In published AML gene-expression datasets, immunoproteasome (IP) subunits PSMB8/LMP7 (p=0.0003***), PSMB9/LMP2 (p=0.0007***) and PSMB10/MECL1 (p & lt;0.0001****) showed significantly higher expression in MLLr compared to non-MLLr-AML. IP is a proteasomal variant constitutively expressed in cells of hematopoietic origin, induced under stimulation with pro-inflammatory cytokines and relevant for mediating stress-responses during inflammation and infection. To assess for functional dependency of MLLr cells on IP subunits we performed an in vitro CRISPR/Cas9 dropout screen in MLLr MOLM-13 cells. Genetic inactivation of PSMB8/LMP7 resulted in outcompetition with 3/5 sgRNAs, while there was less dependency detectable for the other subunits. Specificity of this finding was confirmed in 5 different cell lines (4 MLLr; 1 non-MLLr) by RNAi using 2 shRNAs against PSMB8/LMP7 versus non-targeting control. To confirm these findings in primary cells, we used a previously published conventional LMP7 knockout mouse model (Fehling et al., Science, 1994). LSK cells sorted from the bone marrow (BM) of LMP7 knockout and wildtype mice were retrovirally transformed with either MA9, MLL-ENL (ME) or NUP98-HOXA9 (as non-MLLr control) to assess for disease development by serial plating in methylcellulose. Only in MA9 or ME transformed cells LMP7-deficiency limited re-plating capacity to 2-4 rounds. When we injected 2,5x 104 MA9-infected LSK cells into sublethally irradiated recipient mice, recipients of MA9-LMP7-/- cells (n=12) and MA9-LMP7+/+ (n=12) showed development of AML. However, recipients of MA9-LMP7-/- cells had a significant delay in AML development (median survival 63.0 days for LMP7+/+ versus 92.5 days for LMP7 -/- animals, p=0.0387*). Besides the significant delay in AML development, disease penetrance was reduced by 50%, indicating that deficiency for LMP7 impairs development of MA9 driven AML. In contrast, immunophenotypic abundance of HSPCs in LMP7-/- versus LMP7+/+ animals revealed comparable numbers in all relevant subpopulations. Competitive transplantation of LMP7-/- BM into recipient hosts showed no competitive disadvantage or lack of self-renewal capacity compared to LMP7+/+ controls. Pharmacologic inhibition of IP function using the specific LMP7-inhibitor PR-957 (ONX-0914) resulted in significant delay of disease development in secondary recipient hosts. To assess its effect on LIC frequency we performed limiting dilution assays of MA9 leukemic cells in sublethally irradiated recipient mice. PR-957 treatment reduced LIC frequency compared to DMSO control (1/57410 vs. 1/4450). Pharmacologic inhibition of PSMB8/LMP7 in human MLLr leukemia cell lines induced cellular differentiation. Likewise, cell cycle and metabolism appeared affected, functions which could be confirmed by global transcriptome analysis. Taken together, our studies uncover a selective dependency of MLLr-leukemia on IP function and identify PSMB8/LMP7 as a tractable target. Disclosures Heidel: Celgene: Consultancy; Novartis: Consultancy, Research Funding; CTI: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    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. 140, No. Supplement 1 ( 2022-11-15), p. 491-492
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    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: Experimental Hematology, Elsevier BV, Vol. 124 ( 2023), p. S143-
    Type of Medium: Online Resource
    ISSN: 0301-472X
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 2005403-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Leukemia, Springer Science and Business Media LLC, Vol. 34, No. 5 ( 2020-05), p. 1444-1449
    Type of Medium: Online Resource
    ISSN: 0887-6924 , 1476-5551
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2008023-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Leukemia, Springer Science and Business Media LLC, Vol. 37, No. 10 ( 2023-10), p. 2027-2035
    Abstract: Scribble complex proteins can influence cell fate decisions and self-renewal capacity of hematopoietic cells. While specific cellular functions of Scribble complex members are conserved in mammalian hematopoiesis, they appear to be highly context dependent. Using CRISPR/Cas9-based genetic screening, we have identified Scribble complex-related liabilities in AML including LLGL1 . Despite its reported suppressive function in HSC self-renewal, inactivation of LLGL1 in AML confirms its relevant role for proliferative capacity and development of AML. Its function was conserved in human and murine models of AML and across various genetic backgrounds. Inactivation of LLGL1 results in loss of stemness-associated gene-expression including HoxA -genes and induces a GMP-like phenotype in the leukemia stem cell compartment. Re-expression of HoxA9 facilitates functional and phenotypic rescue. Collectively, these data establish LLGL1 as a specific dependency and putative target in AML and emphasizes its cell-type specific functions.
    Type of Medium: Online Resource
    ISSN: 0887-6924 , 1476-5551
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2023
    detail.hit.zdb_id: 2008023-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Journal of Clinical Investigation, American Society for Clinical Investigation, Vol. 128, No. 10 ( 2018-8-27), p. 4359-4371
    Type of Medium: Online Resource
    ISSN: 0021-9738 , 1558-8238
    Language: English
    Publisher: American Society for Clinical Investigation
    Publication Date: 2018
    detail.hit.zdb_id: 2018375-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 726-727
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    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