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: The FASEB Journal, Wiley, Vol. 20, No. 14 ( 2006-12), p. 2621-2623
    Type of Medium: Online Resource
    ISSN: 0892-6638 , 1530-6860
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2006
    detail.hit.zdb_id: 1468876-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    SAGE Publications ; 2007
    In:  Pediatric and Developmental Pathology Vol. 10, No. 4 ( 2007-08), p. 328-334
    In: Pediatric and Developmental Pathology, SAGE Publications, Vol. 10, No. 4 ( 2007-08), p. 328-334
    Abstract: Achondrogenesis type IA (Houston-Harris) is an extremely rare lethal chondrodysplasia with a characteristic severe disarrangement of endochondral ossification. The growth plate cartilage completely lacks columnar-zone formation and shows chondrocyte expansion due to intracellular vacuoles. This article on a new case of achondrogenesis type IA confirms these findings and demonstrates, on the ultrastructural level, the retention of fine fibrillar material within the rough endoplasmic reticulum (rER). Molecular analysis in the presented case of achondrogenesis type IA did not reveal mutations in the COL2A1 and SLC26A2 genes, which are known to cause achondrogenesis types IB and type II. Although the extracellular cartilage matrix was severely altered, all of the investigated matrix molecules (collagens, aggrecan, matrilins, cartilage oligomeric protein [COMP]) showed a normal distribution pattern. The only exception was type-X collagen, which was significantly reduced. Overall, our study suggests a disturbance in cartilage matrix assembly in the present case due to the retention of some sort of matrix component within the rER. Presumably, as a consequence of this event, processes of chondrocyte maturation and differentiation and endochondral bone formation are severely affected in this case of achondrogenesis type IA.
    Type of Medium: Online Resource
    ISSN: 1093-5266 , 1615-5742
    Language: English
    Publisher: SAGE Publications
    Publication Date: 2007
    detail.hit.zdb_id: 1480654-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Organic Geochemistry, Elsevier BV, Vol. 37, No. 10 ( 2006-10), p. 1289-1302
    Type of Medium: Online Resource
    ISSN: 0146-6380
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2006
    detail.hit.zdb_id: 2018075-5
    detail.hit.zdb_id: 428531-1
    SSG: 13
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Oncology Research and Treatment, S. Karger AG, Vol. 28, No. 2 ( 2005), p. 109-111
    Type of Medium: Online Resource
    ISSN: 2296-5270 , 2296-5262
    Language: English
    Publisher: S. Karger AG
    Publication Date: 2005
    detail.hit.zdb_id: 2749752-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    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 ...
  • 6
    In: Science of The Total Environment, Elsevier BV, Vol. 407, No. 11 ( 2009-5), p. 3459-3472
    Type of Medium: Online Resource
    ISSN: 0048-9697
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2009
    detail.hit.zdb_id: 1498726-0
    detail.hit.zdb_id: 121506-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    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 ...
  • 8
    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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 3249-3249
    Abstract: The human cytomegalovirus (CMV) is an important cause for mortality and morbidity after allogeneic stem cell transplantation (allo SCT) especially in patients without a CMV-specific T cell response. CMV seropositive patients allografted with a CMV seronegative donor do not rise up specific CMV-T cell immunity posttransplant and are therefore at great risk for CMV viremia and disease. In a prospective manner, we analyzed blood samples of 38 HLA-A2+ patients with different haematological malignancies for CMV specific T cells. Methods: Frequency of T cells with reactivity against the pp65-peptide (NLVPMVATV) was assessed by ELISPOT assay in 38 patients at defined time points after allo SCT. In patients with high CMV specific T cell frequencies, the T cell phenotype was determined by flow cytometry. Surveillance of CMV viremia was carried out by routine PCR-technique or pp65 Ag staining. Results: In high-risk patients (donor-/recipient+) viremia was observed in 7/9 patients, 3/7 developed clinical severe CMV-disease. In this group, only one patient presented a weak CMV-specific T cell response in the first year after transplantation, although CMV-specific T cells were demonstrated in 5/9 patients before transplantation. In contrast, 64% (11/17) of the CMV seropositive patients having had a CMV seropositive donor showed CMV-specific T cells around day 30. Their T cell frequencies remained stable at a relative high level during the whole time of our investigation period and no CMV disease was observed. CMV seronegative patients allografted with either a CMV seronegative or seropositive donor also remained disease-free. CMV specific T cells were detectable in 2/7 patients of the d+/r− group. FACS analysis revealed that most of the responding cells were of the activated effector phenotype CD8+/CD45RA+/HLA-DR+ with a low expression of CCR7 and CD27. Conclusion: CMV-seropositive patients receiving graft from a seronegative donor are at a high risk and therefore prone for CMV-viremia and -disease. The development of CMV-specific T cells i.e. immune reconstitution in high risk patients remains delayed or is completely missing during the first year post transplant. In contrast, seropositive recipients grafted with a seropositive donor develop a durable T cell response within 3–4 weeks and show no viremia at all. New strategies as donor vaccination and adoptive T cell transfer are warranted to prevent CMV-disease in CMV seropositive patients for whom only a seronegative donor can be found.
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 3018-3018
    Abstract: There are several biological markers that are now known to confer HR in AML patients as there are cytogenetic high risk aberrations, FLT3 length mutations with a high allelic ratio (AR) and the initial response to therapy as defined by more than 10% blasts in the “day 15” bone marrow blast count (d15). For most of those patients categorized as having HR-AML allogeneic transplantation comprises the only option of cure. Recently we reported (Platzbecker et al., Leukemia 2006) on a highly active transplantation strategy using reduced intensity conditioning that allowed for an upfront allogeneic transplantation in HR-AML patients as part of the induction treatment. In 2003 we started a multicenter randomized trial which investigates both the feasibility and efficacy of allogeneic stem cell transplantation as upfront intensification therapy in HR-AML patients. Here we report the feasibility of the approach. The study is being conducted in a classical 2×2 design with intensified treatment arms B and D that scheduled allogeneic transplantation in HR patients as part of the induction therapy (IT) during marrow aplasia achieved by DA (daunorubicin 60 mg/m2 - day 3–5; cytarabine 100 mg/m2 - day1–7). Intensified risk adapted therapy was compared to a “conventional” treatment strategy (arm A and C) which allows for allogeneic transplantation only in patients achieving remission after two induction courses (DA) with a complex aberrant karyotype or in patients with aberrant chromosome 7 and 5. We identified 109 patients with HR in the intensified treatment groups (B/D) where HR was defined by cytogenetic criteria (n=73); FLT3-AR (n=14) and d15 blast count (n=21). At the same time a fast search strategy was conducted and revealed donor availability (related or unrelated) in 92 patients (84.4%). Consequently, 46 out of 109 HR-AML patients assigned to the intensified treatment strategy (B/D) received allogeneic transplantation within the protocol (42.2%). In contrast, 8 out of 102 HR patients in the none-intensified treatment arms (A/C) were treated with allogeneic transplantation within the protocol (7.8%). Whereas 19 of 109 patients (17%) with an intensified treatment strategy (B/D) were transplanted after achieving remission, 27 of the 109 patients (25%) were treated with allogeneic transplantation during marrow aplasia after IT1 (n=9) or IT2 (n=18) respectively. Available donors were predominantly family donors for patients receiving transplant after IT1 (6/9) but were more often unrelated donors after IT2 (12/18). Allogeneic transplantation was performed in (B/D) HR-AML patients with a median time from diagnosis of the disease to transplantation of 54 days (range 29–204). In conclusion, early allogeneic transplantation is possible in 40–50% of HR-AML patients during the induction phase of therapy in a randomized AML trial. An early upfront transplantation strategy in aplasia is logistically challenging. However, the presented data show that given a fast search strategy at the time point of diagnosis about 25% of patient can be transplanted during the aplasia of induction therapy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    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...