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: Experimental Hematology, Elsevier BV, Vol. 38, No. 8 ( 2010-8), p. 661-665
    Type of Medium: Online Resource
    ISSN: 0301-472X
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2010
    detail.hit.zdb_id: 2005403-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 1751-1751
    Abstract: Abstract 1751 Poster Board I-777 SFRP1 is one of five known secreted frizzled related proteins (SFRPs) whose expression has been recently observed to be down-regulated due to hypermethylation in acute and chronic leukaemia, but so far not in myelodysplastic syndrome (MDS). SFRPs are extracellular antagonists of the Wnt-dependent signalling pathway that plays an important role in the pathogenesis of solid tumours and hematopoietic malignancies. We examined the mRNA expression and the promoter methylation of SFRP1 and the mRNA expression of the activating Wnt membrane receptor frizzled 3 (Fzd3) in bone marrow (BM) mononuclear cells derived from 78 patients with MDS (low risk, n=18; int-1, n=27; int-2, n=19 and high risk, n=14), 23 patients with acute myeloid leukaemia (AML) and 20 patients with acute lymphoblastic leukaemia (ALL) at the time of initial diagnosis as compared to healthy individuals (n=24). We performed real time quantitative RT-PCR to determine mRNA transcription levels of both genes as well as DNA-pyrosequencing to quantify promoter methylation of SFRP1. In addition, we analyzed highly purified CD34+ cells from 45 MDS patients (low risk, n=11; int-1, n=12; int-2, n=11 and high risk, n=11) and 18 healthy controls. In both unselected BM and CD34+ cells of the MDS patients (BM: 15 (83%) of low risk, 23 (85%) of int-1, 17 (89%) of int-2 and 11 (80%) of high risk MDS; CD34+: 10 (91%) low risk, 10 (83%) int-1, 11 (100%) int-2 and 9 (82%) high risk MDS) a significant decreased SFRP1 expression was detected (BM: 3.4 fold for low risk, 2.9 fold for int-1, 7.9 fold for int-2, 15.4 fold for high risk MDS; CD34+: 3.6 fold for low risk, 2.1 fold for int-1, 8.3 fold for int-2 and 9.2 fold for high risk MDS). In selected MDS-patient samples (12.3%) we observed DNA-hypermethylation of the SFRP1 promoter as compared to normal controls. Furthermore, in AML and ALL BM samples, we could confirm previously reported promoter hypermethylation (AML 50% and ALL 61%) and the association with transcriptional down-regulation of SFRP1 (19.8 fold in 22 (95 %) AML patients, 17.3 fold in 17 (85%) ALL patients). In addition, expression levels of Fzd3 were up-regulated in both acute leukaemia and MDS achieving the level of statistical significance in high-and low risk MDS. Our data show a significant transcriptional down-regulation of SFRP1 as a common event in AML, ALL and - as demonstrated for the first time - in MDS. The epigenetic inactivation of SFRP1 and the transcriptional up-regulation of the Wnt receptor Fzd3 seem to be associated with an activation of the disease-related affected Wnt signalling pathway in these hematopoietic diseases. Hypermethylation of the SFRP1 promotor may be one of the predominant regulatory mechanism in AML and ALL, but not mainly in MDS. 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: 2009
    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. 114, No. 22 ( 2009-11-20), p. 2787-2787
    Abstract: Abstract 2787 Poster Board II-763 Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis and an increased risk of evolution to acute myeloid leukemia. The majority of MDS patients will depend on regular transfusions of packed red blood cells (PRBC) during their course of disease due to symptomatic anemia. Since recurrent transfusions of PRBC will result in iron overload with the risk of damage of organs such as heart, endocrine glands and the liver, consequent iron chelation therapy (IC) became an important element of supportive care in MDS patients. Recently, the availability of the oral iron chelator deferasirox provides a convenient management of iron overload in MDS. Since intensive IC has been shown to improve hematopoiesis in iron overloaded patients we performed gene expression profiling on patients with low or intermediate MDS prior and after IC, to elucidate wheter IC leads to alteration of genes involved in hematopiesis, in particular in erythropoiesis. Heparinized bone marrow samples were obtained after informed consent from 6 MDS patients (2 refractory anemia, 4 refractory anemia with ringed sideroblasts) upon initial diagnosis of iron overload (prior IC) and after a period of 1 year of iron chelation (after IC) with the oral iron chelator deferasirox. CD34+ hematopoietic progenitor as well as CD71+ erythroid progenitor cells were isolated by high gradient magnetic cell separation (Miltenyi Biotech, Bergisch Gladbach, Germany). RNA was extracted from CD34+ cells and CD71+ cells using TRIzol reagent (Invitrogen, Life Technologies, Grand Island, NY) according to the manufacturer's protocol. Quality controlled RNA was hybridized according to the standard Affymetrix protocol to HG-U133 Plus 2.0 microarrays. Data analysis was performed using the Gene Spring Software version 4.0 (Silicon genetics, San Carlos, CA). Restrictions were set as follows: only genes that were ‘present' in at least 75% of samples were used for further analyses, genes were considered as ‘differentially expressed' when they showed at least 3 fold change between the different groups. Statistical significance was calculated by non-parametric t-test, with P 〈 0.05. In a first step we compared gene expression patterns of CD71+ cells in MDS patients prior and after IC. In total 106 probe sets representing unique genes, hypthetical proteins and open reading frames matched the restriction settings. In an intensive survey on these genes we identified several genes that have been associated with erythropoiesis including Stromal derived factor-1 (CXCL12), Janus kinase 2 (JAK2), and Heat shock transcription factor 2 (HSF2). To exclude that these changes in gene expression where due to the natural course of the disease in specific patients, we compared gene expression of CD71+ cells from patients after IC to an independent test set of CD71+ MDS samples (n=12). Interestingly, we still found an aberrant expression of these genes, indicating that the observed gene expression changes were related to the IC in these patients rather than to the natural course of diesease. However, we were not able to find an altered expression of these genes in CD34+ progenitor cells prior and after IC, suggesting that the effect on gene expression is restricted to CD71+ cells. Iron overload is an inevitable side effect of regular blood transfusions in MDS patients. Intensive IC has been shown to improve erythropoiesis in iron overloaded patients. We found, that IC results in upregulation of Stromal derived factor-1, Janus kinase 2 and Heat shock transcription factor 2 all of them known to regulate hematopoiesis. Moreover, HSF2 and JAK2 have been closely involved in regulation of erythropoiesis. JAK2 deficiency has been shown to result in abrogated erythropoiesis and therefore increase of JAK2 expression after iron chelation might link IC to improvement of erythropoiesis and subsequently decrease of transfusion requirement in some patients receiving IC. Disclosures: Hofmann: Novartis Oncology, Nürnberg, Germany: 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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 2693-2693
    Abstract: Introduction: Disturbed proliferation and differentiation are the most crucial oncogeneic factors leading to malignant turnover of hematopoiesis in myeloid malignancies. Therefore, estimating the lifetime proliferation status of malignant hematopoietic cells is critical. Recently the hypothesis of an epigenetic molecular clock has been corroborated. Depending on the accumulation of CpG methylation errors throughout life after each cell division it is possible to measure an increased DNA methylation of formerly unmethylated CpG islands and subsequently relate it to the mitotic cell age. In order to elucidate the importance of disturbed proliferation in hematologic diseases we initiated a novel approach for profiling mitotic ages of hematopoietic cells in myelodysplastic syndrome and acute leukemia. Patients & Methods: Bone marrow (BM) cells of patients with myelodysplastic syndrome (MDS, IPSS-low/int-1-risk n=23, IPSS-int-2/high-risk n=27), acute myeloid leukemia (AML, n=55), acute lymphoblastic leukemia (ALL, T-lineage n=40, B-lineage n=8), and of age matched healthy individuals (n=24) were analyzed. In addition, selection of CD34+ cells was performed in MDS (n=43), in AML (n=10) as well as in healthy BM samples (n=31). CD19+ peripheral blood cells from healthy donors (n=13) served as an additional control. Genomic DNA was isolated and bisulfite converted using standard TRIZOL technique (Invitrogen, Carlsbad/CA, USA) followed by EpiTect-Bisulfite-Kit conversion (Qiagen, Hilden, Germany). PCR amplification of a CpG rich 3′ site of the Cardiac Specific Homeobox gene (CSX), considered as an epigenetic molecular clock locus, was performed as previously reported. DNA methylation was quantitative measured using the PyroMark ID Pyrosequencing system (Biotage, Uppsala, Sweden). Quantitative DNA methylation data are presented with mean ± S.E.M. Results: In MDS int-2/high-risk specific DNA methylation of BM (26.6 ± 1.8 %) and CD34+ (28.6 ± 2.7 %) was significant higher compared to low/int-1-risk MDS (BM: 19.2 ± 1.6 %, p=0.0047, CD34+: 18.7 ± 2.4 %, p=0.0093) and healthy donors (BM: 17.8 ± 0.5 %, CD34+: 17.0 ± 0.4 %, p & lt;0.0001). Furthermore, AML BM samples showed significant higher methylation of 34.2 ± 1.7 % compared to MDS BM int-2/high-risk samples (p=0.0081). Interestingly we could detect significant higher differences in CSX methylation between paired BM/CD34+ samples in MDS low/int-1-risk, but not in MDS int-2/high-risk or AML compared to age matched healthy individuals (p=0.0063). Notably, T-lineage ALL samples did show a remarkable high mean methylation of 61.7 ± 3.1 %. However, B-lineage ALL analysis revealed a similar methylation pattern in comparison to healthy CD19+ cells (26.1 ± 1.4 % and 25.1 ± 1.4% respectively). Discussion: The significant higher CSX methylation in AML compared to int-2/high-risk and in int-2/high-risk compared to low/int-1-risk MDS or healthy individuals could possibly be considered as a disease stage related molecular marker. The intra-individual similarity of CSX methylation levels between BM and CD34+ cells in int-2/high-risk MDS patients supports the theory of a stem cell origin of this disease subgroup, whereas low/int-1-risk MDS samples reveal higher differences possibly pointing to an origin in a more differentiated progenitor cell. However, the observation of higher mitotic ages in T-lineage but not B-lineage ALL raises questions about the role of cell proliferation in distinct lymphoblastic leukemias. In summary, the determination of mitotic cell ages by quantitative DNA methylation analysis could contribute to the molecular classification of hematological malignancies and may further be used for riskassessment in patients with MDS.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    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: Leukemia Research, Elsevier BV, Vol. 34, No. 12 ( 2010-12), p. 1610-1616
    Type of Medium: Online Resource
    ISSN: 0145-2126
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2010
    detail.hit.zdb_id: 2008028-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 2781-2781
    Abstract: Abstract 2781 Poster Board II-757 Lenalidomide belongs to a proprietary class of immunmodulatory drugs showing therapeutic activity in patients with myelodysplastic syndrome (MDS), in particular in those having the 5q-abnormality, but also in patients not showing this cytogenetical aberration. In 2008, Ebert et al. (PLos Med. 2, e35) could demonstrate that there is a specific gene expression profile in bone marrow cells collected from MDS-patients either with 5q- syndrome as well as MDS-patients having no 5q-abnormality which is strongly correlated with the clinical response to treatment with lenalidomide. Whereas this finding is not of clinical importance in patients with MDS del 5q (overall response 75 %) it may play a pivotal role for prediction of clinical response to lenalidomide in non-del 5q MDS-patients. Therefore, we have studied gene expression profile (HG-U133plus2.0, Affymetrix, Santa Clara, CA) of routinely isolated low-density mononuclear bone marrow cells from 8 patients with IPSS low/int-1 risk MDS having no deletion on chromosome 5 but were subsequently treated with lenalidomide 5 mg/day. All of the patients were transfusion dependent for red blood cells. The median duration of treatment with lenalidomide was 22 weeks. RNA was extracted by Trizol and quality controlled by using a Bioanalyzer 2100 system (Agilent, Waldborn, Germany) to exclude RNA degradation. Microarray hybridization was performed according to the standard Affymetrix protocol. Data were analyzed by Microarray Analysis Suites 5.0 (MAS 5.0; Affymetrix) and GeneSpring (Agilent Technologies, Santa Clara, CA). For clustering analysis we utilized the gene list of 68 discriminating genes as published by Ebert et al. the molecular analysis did clearly separate two groups of patients having specific gene expression profiles according to the responder/non-responder group as published previously. Furthermore, single sample prediction could discriminate three out of 8 patients to be possible responders to lenalidomide but this was not correlated to the clinical course of those patients while on treatment with lenalidomide. However, none of the MDS-patients receiving lenalidomide did show significant clinical response as defined by reduction of transfusion requirement by 50 % or transfusion independence. In conclusion, prediction of response to lenalidomide in non-del 5q patients by gene expression profiling so far remains critical. Prospective analysis of molecular changes including DNA analysis in larger clinical trials using lenalidomide in non-del 5q MDS-patients are required to establish reliable predictive markers in MDS. Disclosures: Hofmann: Celgene: Research Funding. Platzbecker:Celgene: 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
    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...