GLORIA

GEOMAR Library Ocean Research Information Access

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. 133, No. 21 ( 2019-05-23), p. 2305-2319
    Abstract: Neural cell adhesion molecule 1 (NCAM1; CD56) is expressed in up to 20% of acute myeloid leukemia (AML) patients. NCAM1 is widely used as a marker of minimal residual disease; however, the biological function of NCAM1 in AML remains elusive. In this study, we investigated the impact of NCAM1 expression on leukemogenesis, drug resistance, and its role as a biomarker to guide therapy. Beside t(8;21) leukemia, NCAM1 expression was found in most molecular AML subgroups at highly heterogeneous expression levels. Using complementary genetic strategies, we demonstrated an essential role of NCAM1 in the regulation of cell survival and stress resistance. Perturbation of NCAM1 induced cell death or differentiation and sensitized leukemic blasts toward genotoxic agents in vitro and in vivo. Furthermore, Ncam1 was highly expressed in leukemic progenitor cells in a murine leukemia model, and genetic depletion of Ncam1 prolonged disease latency and significantly reduced leukemia-initiating cells upon serial transplantation. To further analyze the mechanism of the NCAM1-associated phenotype, we performed phosphoproteomics and transcriptomics in different AML cell lines. NCAM1 expression strongly associated with constitutive activation of the MAPK-signaling pathway, regulation of apoptosis, or glycolysis. Pharmacological inhibition of MEK1/2 specifically inhibited proliferation and sensitized NCAM1+ AML cells to chemotherapy. In summary, our data demonstrate that aberrant expression of NCAM1 is involved in the maintenance of leukemic stem cells and confers stress resistance, likely due to activation of the MAPK pathway. Targeting MEK1/2 sensitizes AML blasts to genotoxic agents, indicating a role for NCAM1 as a biomarker to guide AML treatment.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 123, No. 15 ( 2014-04-10), p. 2355-2366
    Abstract: Activating KRAS mutations are detected in a substantial number of hematologic malignancies. In a murine T-cell acute lymphoblastic leukemia (T-ALL) model, we previously showed that expression of oncogenic Kras induced a premalignant state accompanied with an arrest in T-cell differentiation and acquisition of somatic Notch1 mutations. These findings prompted us to investigate whether the expression of oncogenic KRAS directly affects DNA damage repair. Applying divergent, but complementary, genetic approaches, we demonstrate that the expression of KRAS mutants is associated with increased expression of DNA ligase 3α, poly(ADP-ribose) polymerase 1 (PARP1), and X-ray repair cross-complementing protein 1 (XRCC1), all essential components of the error-prone, alternative nonhomologous end-joining (alt-NHEJ) pathway. Functional studies revealed delayed repair kinetics, increased misrepair of DNA double-strand breaks, and the preferential use of microhomologous DNA sequences for end joining. Similar effects were observed in primary murine T-ALL blasts. We further show that KRAS-mutated cells, but not KRAS wild-type cells, rely on the alt-NHEJ repair pathway on genotoxic stress. RNA interference–mediated knockdown of DNA ligase 3α abolished resistance to apoptotic cell death in KRAS-mutated cells. Our data indicate that targeting components of the alt-NHEJ pathway sensitizes KRAS-mutated leukemic cells to standard chemotherapeutics and represents a promising approach for inducing synthetic lethal vulnerability in cells harboring otherwise nondruggable KRAS mutations.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 311-311
    Abstract: Background Acute myeloid leukemia (AML) is a heterogeneous disease of the hematopoietic progenitor cell driven by the subsequent acquisition of genetic alterations. Approximately 20% of AML patients show strong expression of CD56 (neural cell adhesion molecule; NCAM). Expression of NCAM is associated with poor overall survival; however, the functional role of aberrant NCAM expression has not been investigated to date. The goal of this study is to examine the biological role of NCAM in AML and to explore whether NCAM represents a potential therapeutic target. Results In order to evaluate the clinical significance of elevated NCAM expression in AML, we screened a panel of human cell lines for CD56 expression. Most cell lines were positive and cell surface expression correlated with mRNA levels. Knockdown of NCAM with three different doxycycline-inducible shRNAs suppressed cell growth and MTT activity in all positive cell lines. Propidium iodide staining demonstrated an increase in G1 arrest. Importantly, there was a marked apoptosis after NCAM suppression and this effect was proportional to the knockdown efficiency. Survival of NOD-SCIDgamma chain mice in a leukemia engraftment model was significantly prolonged upon NCAM knockdown. Suppression of NCAM sensitized leukemic blasts to treatment with Ara-C or Daunorubicin in vitro and in xenotransplantation experiments. To test the consequences of NCAM overexpression in negative leukemic cell lines we transduced the NCAM transcript from Nomo-1 into HL60 and K562 cells. HL60 cells had a significantly lower sensitivity towards Ara-C or Daunorubicin treatment. IC50 for the BCR-ABL inhibitor Dasatinib in K562 cells increased from 0.95 nM (EV, empty vector) to 2.2 nM in NCAM overexpressing cells. To dissect possible upstream regulation mechanisms of NCAM expression we performed DNAseI hypersensitivity assays coupled to qRT-PCR mapping of known putative sites in the NCAM promoter and observed open chromatin for the binding sites of Meis1, Mef2 and Stat1. shRNA-mediated knockdown of MEIS1, MEF2c and MLL-AF9 resulted in significant suppression of NCAM cell surface expression, suggesting an upstream regulatory role for MLL-AF9. To gain insights into the mechanisms underlying the NCAM function in AML we performed gene expression comparisons of the 30 highest versus 30 lowest expressing samples in the GSE8043 dataset. Fifty-seven Biocharta pathways were differentially expressed between NCAMhigh and NCAMlow samples, while expression changes predicted abnormal cell-cycle regulation, stress and DNA damage response, cell survival, renewal and adhesion. Western blot, protein array and qRT-PCR analyses of candidate downstream signaling pathways upon knockdown of NCAM demonstrated enhanced degradation of BetaCatenin, decreased expression of BCL-2 and increased levels of p21 and p27. The upstream regulation mechanism described above revealed MLL-AF9 (M/A9) as a top candidate for NCAM regulation. Subsequent analysis of M/A9 L-GMPs (Lin- cKit+ CD34+ FcgR+) demonstrated strong surface expression of NCAM, whereas normal HSCs (Lin-cKit+ Sca1+) were NCAM-negative. This could be validated by gene expression analyses of M/A9 L-GMPs compared with normal HSCs. In order to elucidate the role of NCAM on leukemic cell function in a mouse model, NCAM-/- and control wildtype (WT) bone marrow cells were transformed with a retroviral construct of M/A9 and transplanted into lethally irradiated littermates. Recipients of NCAM-/- M/A9 cells developed acute leukemia with prolonged disease latency. NCAM-/- M/A9 cells had lower CD117 and Gr-1 expression, but higher expression of Mac-1 and, in some samples, aberrant B220 co-expression. Importantly, there was a reduced representation of L-GMPs in the NCAM-/- M/A9 group and limited dilution retransplantation assays revealed a significantly prolonged survival of NCAM-/- M/A9 mice. Replating activity in methylcellulose was diminished and could be eradicated with sublethal doses of Cytarabine. Summary Targeting aberrant expression of NCAM demonstrated strong antileukemic activity in vitro and sensitized leukemic blasts to genotoxic stress. In vivo, depletion of NCAM resulted in prolonged disease survival in syngeneic and xenotransplantation experiments and diminished self-renewal capacities. Our data suggest that NCAM represents a promising therapeutic strategy and likely targets AML cells at the LSC level. 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: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 3789-3789
    Abstract: SIRT1 is a NAD+ dependent histone deacetylase, which has been shown to act as an important regulator of apoptosis, DNA-repair and is involved in the maintenance of genetic integrity under conditions of cellular stress. Beside deacetylation of histones H4K16, SIRT1 has numeral other substrates including KU70, FOXO1 or p53. SIRT1 deacetylates p53 at lysine 382 thereby reducing its transcriptional activity followed by loss of p53 dependent apoptosis in response to cell damage. The activity of SIRT1 is negatively regulated by DBC1 (Deleted in Breast Cancer 1) and involves protein–protein interaction (Kim et al., Nature 2008). Recent reports have demonstrated increased expression of SIRT1 in leukemic cells. Of note, SIRT1 seems to be specifically essential to maintain stem cell activity in CML leukemic stem cells (Li et al, Cancer Cell, 2012), however the role of SIRT1 in acute myeloid leukemia (AML) and in the context of other oncogenic tyrosine kinases is poorly understood. Aims To characterize the role of SIRT1 in acute myeloid leukemia in the context of defined genetic backgrounds. Methods To investigate the functional role of SIRT1 we performed expression analysis studies. Inhibition of SIRT1 was achieved by shRNA experiments and pharmacological targeting. Functional consequences were addressed by immunoblotting, co-immunoprecipitation, cell death assays, gamma-H2AX staining and xenotransplantion experiments. Results SIRT1 protein expression analysis in primary human AML samples (n=20) and cell lines (n=13) showed preferential high expression levels in cells harboring FLT3-ITD mutations or oncogenic KRAS/NRAS. Inhibition of FLT3 signaling using shRNA-mediated knockdown of FLT3 or upon treatment with tyrosine kinase inhibitors caused a time and dose dependent suppression of SIRT1 expression accompanied by an increase in p53 acetylation. Vice versa, overexpression of FLT3-ITD in BaF/3 cells or in several murine leukemia models (e.g. MLL/AF9 +/-FLT3-ITD; AML1/ETO +/-FLT3-ITD) induced significant upregulation of SIRT1. Targeting SIRT1 using the SIRT1/2 inhibitor Tenovin-6 (TV-6) or knockdown of SIRT1 resulted in a slight increase in apoptotic cell death in primary AML samples and cell lines. In contrast, inhibition of SIRT1 significantly sensitized to FLT3 inhibitor therapy. Of note, the combination of TV-6 and midostaurin inhibited colony growth and replating activity in MLL-AF9 or AML1-ETO cells harboring FLT3-ITD mutations. Further, knockdown of SIRT1 in the AML cell line MV4-11 expressing a doxycycline-regulated SIRT-shRNA resulted in prolonged survival in a xenotransplantation model. The observed effects prompted us to investigate whether mutated FLT3 is directly involved in the regulation of SIRT1. Inhibition of FLT3-ITD kinase activity caused an increased phosphorylation of the physiological SIRT1-inhibitor DBC1 followed by binding of SIRT1 as revealed by co-immunoprecipitation experiments. We also noticed a slight increase in ATM/ATR phosphorylation and an increase in ATM-DBC1 binding upon FLT3-ITD inhibition indicating SIRT1 inhibition via the ATM-DBC1-SIRT1 axis. These effects were substantially enhanced upon treatment with genotoxic agents (cytarabine; irradiation) in combination with midostaurin compared to either agent alone and were accompanied by increased p53 acetylation and an increased number of gamma-H2AX foci. Conclusion SIRT1 is overexpressed in AML cells and partially regulated by FLT3-ITD or oncogenic RAS. In FLT3-ITD mutated cells SIRT1 is constitutively activated, likely via the inhibition of the physiological inhibitor DBC1. Pharmacologic targeting of SIRT1 sensitizes AML cells to TKI therapy or genotoxic agents and partially restored the FLT3-ITD associated defective stress response pathway ATM-DBC1-SIRT1 resulting in activation of p53. Disclosures: Kindler: Novartis: Membership on an entity’s Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 5223-5223
    Abstract: STAT5 proteins are transcription factors involved in the regulation of proliferation, apoptosis and differentiation. Upon ligand-mediated activation, phosphorylated STAT5A (pSTAT5A) and pSTAT5B form homo- or heterodimers, translocate to the nucleus and activate transcription. Recent data indicate an important role of un-phosphorylated STAT5 (uSTAT5) in the regulation of differentiation. For example, the presence of uStat5 shifts the balance toward the maintenance of leukemic stem cells, whereas genetic depletion of Stat5induced differentiation. Further, uSTAT5 has been shown to act as a repressor of megakaryocytic differentiation via restricting the access of ERG to its target genes. To further investigate the function of uSTAT5 in mammalian AML, we established several cell line models expressing doxycycline-inducible shRNA directed against either STAT5A or STAT5B. In uSTAT5-expressing AML cells targeting of STAT5A or STAT5B suppressed cell growth and induced differentiation. These effects were significantly stronger upon knockdown of STAT5B compared to STAT5A. Global RNA sequencing demonstrated enrichment of differentiation programs upon downregulation of uSTAT5B. To further explore the biological function of uSTAT5, we performed SILAC-based global proteomics after pulldown of either STAT5A or STAT5B. While uSTAT5A was found to be associated with proteins involved in RNA processing, uSTAT5B primarily co-precipitated with chromatin- and histone-binding proteins, like the transcriptional repressor ETV6 or the histone H3K4 demethylase KDM5C. To dissect the specific roles of pSTAT5B and uSTAT5B, we performed co-immunoprecipitation assay upon treatment of AML cells with GM-CSF or vehicle control. As expected, GM-CSF treatment caused strong phosphorylation of STAT5A/B, however, uSTAT5B-binding of ETV6 was almost completely abolished, indicating a specific association only with uSTAT5B. As depletion of uSTAT5B induced differentiation and uSTAT5B interacts with KDM5C and ETV6, we hypothesized that uSTAT5B prevents transcription of genes involved in differentiation. To address this question, we performed H3K4me3 ChIP-seq analysis in THP-1 cells upon knockdown of STAT5B. Downregulation of uSTAT5B caused strong accumulation of H3K4me3 at promoter regions of previously silenced genes. Finally, we wanted to investigate whether targeting of KDM5C can reverse disturbed differentiation in AML cells. Treatment with the KDM5 inhibitor CPI-455 significantly inhibited proliferation and induced apoptosis, but only in AML samples expressing uSTAT5B. In summary, our data provide evidence that uSTAT5B specifically blocks differentiation in AML cells via its interaction with KDM5C followed by repression of H3K4me3 at distinct promoter regions. Targeting of uSTAT5B or its interacting partners might represent an interesting novel strategy in AML therapy. Citation Format: Jakub Szybinski, Daniel Sasca, Jan Heidelberger, Karolin Klumb, Viral Shah, Anna Dolnik, Matthias Theobald, Lars Bullinger, Petra Beli, Thomas Kindler. Epigenetic silencing mediated by non-phosphorylated STAT5B prevents differentiation in acute myeloid leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 5223.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Haematologica, Ferrata Storti Foundation (Haematologica), Vol. 108, No. 10 ( 2023-04-27), p. 2837-2843
    Type of Medium: Online Resource
    ISSN: 1592-8721 , 0390-6078
    Language: Unknown
    Publisher: Ferrata Storti Foundation (Haematologica)
    Publication Date: 2023
    detail.hit.zdb_id: 2186022-1
    detail.hit.zdb_id: 2030158-3
    detail.hit.zdb_id: 2805244-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 1 ( 2013-01-01), p. 373-384
    Abstract: MOZ-TIF2 is a leukemogenic fusion oncoprotein that confers self-renewal capability to hematopoietic progenitor cells and induces acute myelogenous leukemia (AML) with long latency in bone marrow transplantation assays. Here, we report that FLT3-ITD transforms hematopoietic cells in cooperation with MOZ-TIF2 in vitro and in vivo. Coexpression of FLT3-ITD confers growth factor independent survival/proliferation, shortens disease latency, and results in an increase in the number of leukemic stem cells (LSC). We show that STAT5, a major effector of aberrant FLT3-ITD signal transduction, is both necessary and sufficient for this cooperative effect. In addition, STAT5 signaling is essential for MOZ-TIF2–induced leukemic transformation itself. Lack of STAT5 in fetal liver cells caused rapid differentiation and loss of replating capacity of MOZ-TIF2–transduced cells enriched for LSCs. Furthermore, mice serially transplanted with Stat5−/− MOZ-TIF2 leukemic cells develop AML with longer disease latency and finally incomplete penetrance when compared with mice transplanted with Stat5+/+ MOZ-TIF2 leukemic cells. These data suggest that STAT5AB is required for the self-renewal of LSCs and represents a combined signaling node of FLT3-ITD and MOZ-TIF2 driven leukemogenesis. Therefore, targeting aberrantly activated STAT5 or rewired downstream signaling pathways may be a promising therapeutic option. Cancer Res; 73(1); 373–84. ©2012 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 136, No. 21 ( 2020-11-19), p. 2442-2456
    Abstract: The interaction of menin (MEN1) and MLL (MLL1, KMT2A) is a dependency and provides a potential opportunity for treatment of NPM1-mutant (NPM1mut) and MLL-rearranged (MLL-r) leukemias. Concomitant activating driver mutations in the gene encoding the tyrosine kinase FLT3 occur in both leukemias and are particularly common in the NPM1mut subtype. In this study, transcriptional profiling after pharmacological inhibition of the menin-MLL complex revealed specific changes in gene expression, with downregulation of the MEIS1 transcription factor and its transcriptional target gene FLT3 being the most pronounced. Combining menin-MLL inhibition with specific small-molecule kinase inhibitors of FLT3 phosphorylation resulted in a significantly superior reduction of phosphorylated FLT3 and transcriptional suppression of genes downstream of FLT3 signaling. The drug combination induced synergistic inhibition of proliferation, as well as enhanced apoptosis, compared with single-drug treatment in models of human and murine NPM1mut and MLL-r leukemias harboring an FLT3 mutation. Primary acute myeloid leukemia (AML) cells harvested from patients with NPM1mutFLT3mut AML showed significantly better responses to combined menin and FLT3 inhibition than to single-drug or vehicle control treatment, whereas AML cells with wild-type NPM1, MLL, and FLT3 were not affected by either of the 2 drugs. In vivo treatment of leukemic animals with MLL-r FLT3mut leukemia reduced leukemia burden significantly and prolonged survival compared with results in the single-drug and vehicle control groups. Our data suggest that combined menin-MLL and FLT3 inhibition represents a novel and promising therapeutic strategy for patients with NPM1mut or MLL-r leukemia and concurrent FLT3 mutation.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 124, No. 1 ( 2014-07-03), p. 121-133
    Abstract: SIRT1 is highly expressed in subsets of patients with acute myeloid leukemia harboring activating mutations in signaling pathways and is regulated at the protein levels. Targeting SIRT1 sensitizes leukemic blast to tyrosine kinase inhibitor treatment or chemotherapy via restoration of p53 activity.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 6226-6227
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...