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: British Journal of Haematology, Wiley, Vol. 110, No. 3 ( 2000-09), p. 537-546
    Type of Medium: Online Resource
    ISSN: 0007-1048
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2000
    detail.hit.zdb_id: 1475751-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 25, No. 30 ( 2007-10-20), p. 4813-4820
    Abstract: Applying current diagnostic methods, overt CNS involvement is a rare event in childhood acute lymphoblastic leukemia (ALL). In contrast, CNS-directed therapy is essential for all patients with ALL because without it, the majority of patients eventually will experience relapse. To approach this discrepancy and to explore potential distinct biologic properties of leukemic cells that migrate into the CNS, we compared gene expression profiles of childhood ALL patients with initial CNS involvement with the profiles of CNS-negative patients. Patients and Methods We evaluated leukemic gene expression profiles from the bone marrow of 17 CNS-positive patients and 26 CNS-negative patients who were frequency matched for risk factors associated with CNS involvement. Results were confirmed by real-time quantitative polymerase chain reaction analysis and validated using independent patient samples. Results Interleukin-15 (IL-15) expression was consistently upregulated in leukemic cells of CNS-positive patients compared with CNS-negative patients. In multivariate analysis, IL-15 expression levels greater than the median were associated with CNS involvement compared with expression equal to or less than the median (odds ratio [OR] = 10.70; 95% CI, 2.95 to 38.81). Diagnostic likelihood ratios for CNS positivity were 0.09 (95% CI, 0.01 to 0.65) for the first and 6.93 (95% CI, 2.55 to 18.83) for the fourth IL-15 expression quartiles. In patients who were CNS negative at diagnosis, IL-15 levels greater than the median were associated with subsequent CNS relapse compared with expression equal to or less than the median (OR = 13.80; 95% CI, 3.38 to 56.31). Conclusion Quantification of leukemic IL-15 expression at diagnosis predicts CNS status and could be a new tool to further tailor CNS-directed therapy in childhood ALL.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2007
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Nature Medicine, Springer Science and Business Media LLC, Vol. 12, No. 10 ( 2006-10-01), p. 1191-1197
    Type of Medium: Online Resource
    ISSN: 1078-8956 , 1546-170X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2006
    detail.hit.zdb_id: 1484517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 1456-1456
    Abstract: Severe congenital neutropenia (SCN; Kostmann syndrome) is characterized by a maturation block in the myelopoiesis at the promyelocytic/myelocytic stage leading to decreased amounts of neutrophils in bone marrow and peripheral blood. This maturation arrest was previously associated with an accelerated apoptosis. Administration of recombinant human granulocyte colony-stimulating factor (G-CSF) sufficiently increases neutrophil numbers in most patients. Recently, it was demonstrated that the elevated degree of apoptosis was accompanied by a selective decreased expression of the anti-apoptotic Bcl-2 protein in myeloid progenitor cells in 4 surviving members of the original “Kostmann family” and 1 patient with SCN of unknown inheritance (Carlsson et al., Blood2004; 103: 3355–3361). In these patients, it was shown that the administration of G-CSF restored Bcl-2 expression and corrected the abnormal acceleration of apoptosis. To test whether this observation is a common feature of SCN patients myeloid progenitor cells, we analyzed the mRNA and protein expression of bcl-2 in CD33+ bone marrow myeloid progenitor cells from 6 G-CSF treated SCN patients not related to the original “Kostmann family” in comparison to that of 2 long-term G-CSF treated patients with neutropenia other than SCN and healthy controls without and after administration of G-CSF (4 and 3, respectively). Additionally, the mRNA expression of the Bcl-2-related genes, bcl-xL (anti-apoptotic) and bax (pro-apoptotic), and the expression of caspase 9, an important intracellular amplifier of apoptotic signaling was investigated. Gene expression was measured by quantitative real-time PCR, and protein expression by immunofluorescence and confocal microscopy. We observed a significant increase of bcl-2 expression after administration of G-CSF in CD33+ cells of healthy controls (fold change (FC) = 1.7, p = 0.0011, t-test). In contrast, bcl-2 was significantly lower expressed in CD33+ cells from long-term G-CSF treated SCN patients as compared to that of long-term G-CSF treated patients with neutropenia other than SCN (FC = 20, p = 0.015, t-test), and healthy controls without (FC = 7, p 〈 0.0001) and after administration of G-CSF (FC = 13, p 〈 0.0001). The expression of bcl-xL did show a similar pattern with a significant difference comparing SCN patients and healthy controls without G-CSF (FC = 4.4, p = 0.018). In contrast, the expression of caspase 9 was significantly upregulated in CD33+ cells of SCN patients as well as G-CSF treated controls compared to healthy controls without G-CSF administration (FC = 8.9, p = 0.002). Bax was similarly expressed in all groups. The expression pattern of bcl-2 was confirmed on the protein level. In summary, the expression of bcl-2 was defective in SCN patients as it was described by Carlsson et al., and a similar pattern was observed for bcl-xL. In contrast to published data, long-time administration of G-CSF did not normalize the expression of both anti-apoptotic Bcl-2 family members in our patients. We conclude, that a mechanism other than the restoration of Bcl-2 and Bcl-xL expression is responsible for the increase of neutrophils after G-CSF treatment in our patient sample.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-01), p. 2270-2270
    Abstract: Targeted but risk-adapted therapy to the central nervous system (CNS) has become a prerequisite for successful treatment of childhood acute lymphoblastic leukemia (ALL). Overt CNS involvement is a rare observation in childhood ALL and commonly associated with a poorer outcome when compared to CNS-negative patients. Although there are some known risk factors for CNS involvement of ALL such as T-cell-precursor immunophenotype and high initial white blood cell (WBC) count, the underlying biology leading to CNS disease is poorly understood. We hypothesized that leukemic cells crossing the blood-brain barrier early have distinct biological properties which are reflected in a specific gene expression pattern. To test this hypothesis, we generated leukemic gene expression profiles of 17 patients with CNS involvement using 26 CNS-negative patients as controls. Microarrays containing more than 39,000 distinct cDNA clones (SFGF, Stanford CA) were used. Data were analyzed applying Significance Analysis of Microarrays (PNAS2001, 98, 5116–5121). Our analytical approach identified several differentially expressed genes. To control for possible T-cell contamination of the samples, we purified leukemic cells from four samples and repeated the analysis. Interleukin 15 (IL-15) was one of the genes for which differential expression could be confirmed with an up to 10-fold higher expression in CNS-positive patients. IL-15 is a pro-inflammatory cytokine known to stimulate proliferation and activation of T-lymphocytes and polymorphonuclear cells. Of importance with regard to the CNS, IL-15 was described to be highly expressed in peripheral blood (PBL) mononuclear cells in multiple sclerosis. The high expression of IL-15 in patients with CNS involvement was confirmed in an independent set of 13 CNS-positive and 26 CNS-negative patients by quantitative RT-PCR in a similar range as described above (P Mann-Whitney U-Test = 〈 0.001). Therefore, we postulated that inflammatory processes might be involved in the pathogenesis of CNS disease by increasing the permeability of the blood-brain-barrier and reviewed differential PBL smears of 134 CNS-positive and 335 CNS-negative ALL patients. We observed a significantly higher ratio of immature to total myeloid cells in CNS-positive samples (P = 〈 0.001). Next, we compared IL-15 expression at initial diagnosis of CNS-negative patients subsequently relapsing with CNS involvement (n = 22) to those without CNS disease and being in long-term remission (n = 18). Again, a significantly higher IL-15 expression was detected in patients with CNS relapse (P = 0.01). We conclude that measurement of IL-15 expression could serve as an additional tool to further tailor CNS-directed therapy in children newly diagnosed with ALL. Our results support a role for inflammatory processes, but additional studies are needed to finally elucidate the role of IL-15 and inflammation in the pathogenesis of CNS disease in childhood ALL.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: BMC Medical Genetics, Springer Science and Business Media LLC, Vol. 6, No. 1 ( 2005-12)
    Type of Medium: Online Resource
    ISSN: 1471-2350
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2005
    detail.hit.zdb_id: 2041359-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 500-500
    Abstract: Lymphoid enhancer-binding factor 1 (LEF-1) belongs to the canonical Wnt signaling pathway acting in transcriptional complexes with β-catenin. LEF-1 can also act independent of β-catenin (i.e. in the TGF-β or Notch pathway). Additionally, recent studies described LEF-1 dominant negative isoform (dnLEF-1), which lack the β-catenin binding domain and functions as either a transcriptional repressor or activator. To date, analysis of the role of LEF-1 in hematopoiesis has been restricted to the lymphoid compartment. Previously we described the crucial role of lymphoid enhancer-binding factor 1 (LEF-1) in granulopoiesis. We found that LEF-1 mediates proliferation, survival, and differentiation of granulocyte progenitor cells. Moreover, abrogated LEF-1 expression is one of the pathomechanism of severe congenital neutropenia CN (Skokowa et al., Nature Medicine, in press). Based on these findings, we aimed to characterize the molecular mechanisms of LEF-1 in the regulation of granulocytic differentiation. C/EBPα is well known as a key transcription factor in granulopoiesis and we found it to be a target gene directly regulated by LEF-1. A screen of the known 566 bp upstream promoter of C/EBPα gene revealed a putative LEF-1 binding site (− 559 bp to − 538 bp). We confirmed LEF-1 binding to C/EBPα promoter in nuclear extracts from CD34+ and CD33+ cells in the transcription factor binding NoShift and ChIP assays. Interestingly, LEF-1 binds to the C/EBPα promoter more efficiently in CD33+ myeloid progenitors than in CD34+ cells. The direct regulation of C/EBPα by LEF-1 was further confirmed in functional studies. We found that in line with down-regulation of LEF-1, expression of C/EBPα was also significantly reduced in CD33+ myeloid progenitors of CN patients. Moreover, LEF-1 rescue of these cells resulted in a marked up-regulation of C/EBPα mRNA expression and in vitro restoration of defective granulocytic differentiation. Remarkably, transduction of CN CD33+ cells with dnLEF-1 isoform, which lacks the ß-catenin-binding domain, resulted in up-regulation of C/EBPα to a similar degree as it was observed with full-length LEF-1. A direct regulatory link between LEF-1 and C/EBPα was additionally confirmed in LEF-1 inhibition experiments. C/EBPα expression was significantly down-regulated in CD34+ cells of healthy individuals, transduced with LEF-1 shRNA. Therefore, we propose that LEF-1 is a key regulator of myeloid differentiation acting in a β-catenin-independent manner, similar as it is known for LEF-1 regulation of T-lymphocyte development.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 387-387
    Abstract: Pre-B-cell colony-enhancing factor (PBEF) was first isolated from blood lymphocytes and was found to be involved in the maturation of B-cell precursors. Recently, PBEF was described as cytosolic enzyme nicotinamide phosphoribosyltranserase (NAmPRTase), promoted NAD+ biosynthesis. In this study we tested the ability of PBEF to induce myelopoiesis. Primary CD34+ cells treated with 50 ng/ml of PBEF in the serum-free RPMI medium without any additional cytokines, led to differentiation into mature granulocytes and macrophages, as assessed by FACS analysis (expression of CD11b, CD14, CD16), and by morphology. The same results were observed after lentiviral transduction of CD34+ progenitors with PBEF-GFP reporter. PBEF-dependent maturation was accompanied by increased mRNA expression of hematopoietic transcription factors such as C/EBPα , C/EBPε , PU.1, and ELA2. Moreover, PBEF had synergistic effect on G-CSF-triggered myeloid differentiation of CD34+ cells. Interestingly, G-CSF-treated differentiated CD34+ cells produced increased amounts of PBEF mRNA, as well as of intracellular and secreted PBEF protein in a time-dependent manner, as measured by real-time RT-PCR, ELISA, FACS analysis, and confocal microscopy. Increased synthesis of PBEF was in line with high NAD+ levels in G-CSF-treated cells. Expression of PBEF mRNA during myeloid differentiation was measured by laser-assisted single-cell picking and real-time quantitative RT-PCR analysis of different myeloid precursor cells (myeloblasts, promyelocytes, myelocytes, metamyelocytes, mature granulocytes and monocytes) from bone marrow smears of healthy individuals. PBEF mRNA expression was continually increased during myeloid differentiation with highest levels in mature granulocytes and monocytes. In addition, we compared expression patterns of PBEF mRNA/protein in CD33+ progenitors and PMNs of healthy individuals after in vivo and in vitro treatment with G-CSF. We found PBEF mRNA/protein expression to be markedly increased in both cell types after G-CSF simulation (12 times for granulocytes, and 10 times for monocytes). Intriguingly, PBEF mRNA/protein expression in CD33+ cells and PMNs from G-CSF-treated patients with severe congenital neutropenia (CN) was also up-regulated and was significantly higher, as in G-CSF-treated healthy donors, suggesting that myeloid cells from CN patients try to overcome the differentiation arrest by upregulating PBEF. This was also supported by the fact that CN patients had at least 20 times higher PBEF plasma levels, in comparison to control group. G-CSF-dependent increase of PBEF expression was in line with increased NAD+ levels in G-CSF-treated cells. Indeed, in vitro PBEF treatment as well as lentiviral transduction of CD34+ cells from one CN patient with PBEF-GFP reporter gene resulted in partial restoration of myelopoiesis in vitro. Taken together, PBEF promotes maturation of myeloid cells by NAD+ dependent pathway. Moreover, increased expression of PBEF with subsequent up-regulation of NAD+ synthesis in CD33+ cells and neutrophils from CN patients could reveal mechanisms of G-CSF-dependent restoration of defective myelopoiesis in these patients. rPBEF protein and anti-PBEF antibody were provided by Amgen, USA
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Nature Medicine, Springer Science and Business Media LLC, Vol. 15, No. 2 ( 2009-2), p. 151-158
    Type of Medium: Online Resource
    ISSN: 1078-8956 , 1546-170X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2009
    detail.hit.zdb_id: 1484517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Nature Medicine, Springer Science and Business Media LLC, Vol. 12, No. 11 ( 2006-11), p. 1329-1329
    Type of Medium: Online Resource
    ISSN: 1078-8956 , 1546-170X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2006
    detail.hit.zdb_id: 1484517-9
    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...