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. 121, No. 1 ( 2013-01-03), p. 178-187
    Abstract: The origin of aberrant DNA methylation in cancer remains largely unknown. In the present study, we elucidated the DNA methylome in primary acute promyelocytic leukemia (APL) and the role of promyelocytic leukemia–retinoic acid receptor α (PML-RARα) in establishing these patterns. Cells from APL patients showed increased genome-wide DNA methylation with higher variability than healthy CD34+ cells, promyelocytes, and remission BM cells. A core set of differentially methylated regions in APL was identified. Age at diagnosis, Sanz score, and Flt3-mutation status characterized methylation subtypes. Transcription factor–binding sites (eg, the c-myc–binding sites) were associated with low methylation. However, SUZ12- and REST-binding sites identified in embryonic stem cells were preferentially DNA hypermethylated in APL cells. Unexpectedly, PML-RARα–binding sites were also protected from aberrant DNA methylation in APL cells. Consistent with this, myeloid cells from preleukemic PML-RARα knock-in mice did not show altered DNA methylation and the expression of PML-RARα in hematopoietic progenitor cells prevented differentiation without affecting DNA methylation. Treatment of APL blasts with all-trans retinoic acid also did not result in immediate DNA methylation changes. The results of the present study suggest that aberrant DNA methylation is associated with leukemia phenotype but is not required for PML-RARα–mediated initiation of leukemogenesis.
    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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 398-398
    Abstract: Abstract 398 DNA methyltransferases (DNMT) play an important role in regulation of DNA methylation and mutations of DNMT3A are frequently found in AML. We analyzed the effects of DNMT3B overexpression on leukemogenesis using a tetracycline-inducible DNMT3B mouse model. To analyze the impact of DNMT3B overexpression on leukemogenesis, we retrovirally co- transduced lineage-negative bone marrow cells of wildtype and DNMT3B transgenic mice with an MSCV- cMyc-bcl2 or MSCV-MLL-AF9-hCD4 and an MSCV- tTA- GFP expressing vector. Using these conditions, doxycycline suppressed DNMT3B expression whereas absence of doxycycline led to overexpression of DNMT3B on the mRNA and protein level. To verify that expression of DNMT3b in this mouse model was able to induce DNA methylation, we performed Reduced Representation Bisulfite Sequencing (RRBS) on cMyc-Bcl2 leukemic bone marrow cells. Analysis of the global methylation levels revealed a strong hypermethylation in DNMT3b overexpressing bone marrow cells. Differentially methylated regions (DMR) were defined as those regions with at least 30% difference in methylation levels. DMRs were mainly located in promoter regions and first introns and the heatmap of these DMRs confirms distinct hypermethylation in DNMT3b transgenic cells whereas very few regions become hypomethylated in transgenic cells when compared to wildtype cells. This specific methylation pattern could be responsible for altered gene expression and consecutively altered (stem) cell characteristics. To analyze the influence of changed DNA methylation levels induced by expression of DNMT3b on leukemogenesis, sorted GFP+cells were transplanted into sublethally irradiated wildtype recipients. Mice with and without DNMT3B expression developed leukemia with splenomegaly and a tendency of delayed leukemogenesis in DNMT3B overexpressing mice. Mice with cMyc-Bcl2- and MLL-AF9-induced leukemia showed a clear myeloid phenotype with enhanced expression of the stem cell marker c-kit on MLL-AF9 leukemic cells. To consider the leukemogenic capacity, we transplanted the leukemic blasts into secondary recipients. cMyc-Bcl2 induced leukemia of both, wildtype and DNMT3b recipients was transplantable and lethal. DNMT3B tg leukemic cells were severely impaired in leukemia development in secondary recipients. Secondary recipients of cMyc-Bcl2 induced leukemic DNMT3B cells died significantly later (p= 0.04). As differential methylation can also have an effect on the responsiveness to chemotherapy, spleen cells from MLL-AF9 leukemic mice were treated with different cytotoxic and demethylating drugs to see if DNMT3b induced hypermethylation confers a resistance or increased sensitivity to chemotherapeutic treatment. DNMT3b overexpressing cells were more sensitive to treatment with Azacytidine and Decitabine. Taken together, these findings demonstrate that expression of DNMT3b in a doxycycline-regulatable mouse model induces widespread DNA hypermethylation, whereas a few small regions become hypomethylated when compared to wildtype cells. This specific methylation pattern is associated with delayed leukemogenesis and DNMT3b expression significantly impaired leukemia maintenance. In addition, response to chemotherapeutic treatment is altered by DNMT3b induced hypermethylation which underlines the importance of epigenetic regulatory mechanisms as promising targets for new therapeutic strategies in AML. Disclosures: Müller-Tidow: Celgene: Lecture fees Other; Deutsche Forschungsgemeinschaft: 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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Experimental Hematology, Elsevier BV, Vol. 43, No. 9 ( 2015-09), p. 794-802.e4
    Type of Medium: Online Resource
    ISSN: 0301-472X
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2015
    detail.hit.zdb_id: 2005403-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 127, No. 12 ( 2016-03-24), p. 1575-1586
    Abstract: Increased gene body methylation inhibits leukemia, and oncogenes require varying levels of DNA methylation for efficient leukemogenesis. Dnmt3b-induced DNA methylation in mice targets stem cell–associated genes with prognostic association in acute myeloid leukemia patients.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 20, No. 4 ( 2014-02-15), p. 814-826
    Abstract: Purpose: Cancer cell phenotypes are partially determined by epigenetic specifications, such as DNA methylation. Metastasis development is a late event in cancerogenesis and might be associated with epigenetic alterations. Experimental Design: An in vivo selection approach was used to generate highly aggressive non–small cell lung cancer (NSCLC) cell lines (A549 and HTB56) followed by genome-wide DNA methylation analysis. Furthermore, the therapeutic effects of the epigenetic agent azacytidine on DNA methylation patterns and the in vivo phenotypes were explored. Results: Widespread changes of DNA methylation were observed during development of highly aggressive cell lines. Up to 2.5% of the CpG-rich region was differentially methylated as identified by reduced representation bisulfite sequencing compared with the less aggressive parental cell lines. DNA methyltransferase inhibition by azacytidine reversed the prometastatic phenotype; this was highly associated with the preferential loss of DNA methylation at sites that were hypermethylated during the in vivo selection. Of note, polycomb (PRC2) binding sites were particularly affected by DNA methylation changes after azacytidine exposure that persisted over time. Conclusions: We could show that metastatic capability of NSCLC is closely associated with DNA methylome alterations. Because inhibition of DNA methyltransferase reversed metastasis-prone phenotype, epigenetic modulation seems to be a potential therapeutic approach to prevent metastasis formation. Clin Cancer Res; 20(4); 814–26. ©2013 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2436-2436
    Abstract: Abstract 2436 The origin of altered DNA methylation in cancer and more specifically in leukemia remains incompletely understood. One favored hypothesis suggests that aberrant methylation is recruited to direct genomic target regions of transcriptional repressor oncogenes. PML-RARα, as a transcriptional repressor oncogene, mediates a repressed chromatin structure at its genomic targets and leads to a rather uniform leukemia subtype. In the current study we aimed to identify patterns and variability of DNA methylation changes in PML-RARα associated acute promyelocytic leukemia (APL). We used reduced representation bisulfite sequencing to investigate a large number of samples at single CpG-site resolution with a focus on CpG islands and related promoters. Overall, 20 APL patient samples were sequenced from patients at primary diagnosis. As controls we used 10 remission bone marrow samples and three sets of MACS-purified CD34+ hematopoetic progenitor cells from healthy donors as well as in vitro differentiated promyelocytes. Using Illumina next generation sequencing we obtained about 1 × 107 uniquely mapped bisulfite sequencing reads per patient in total. More than 10 sequencing reads were available for on average 1.2 million distinct CpG sites per sample. Unsupervised hierarchical clustering revealed that APL samples clustered separately from remission bone marrow, CD34+ progenitors and promyelocytes. Using Poisson-Regression (p 〈 10−5) and at least a 2-fold change in methylation as threshold for analysis, primary diagnosis samples where markedly hypomethylated versus controls. 2160 promoter regions were more than 2-fold hypomethylated in primary diagnosis samples compared to controls. However, 716 promoter regions were also 2-fold hypermethylated. A microarray data analysis (www.leukemia-gene-atlas.org) was carried out. The top hypomethylated genes, as sorted by fold changes in methylation and p-values, showed differential expression. Using published PML-RARα binding sites, we also examined methylation differences within these. A total of 11026 CpG-sites comprising 368 promoters could be taken into account. Notably, there was a high degree of hypomethylation at PML-RARα binding sites when comparing primary diagnosis samples with controls, while only a few binding sites appeared hypermethylated. Differential hypomethylation at PML-RARα binding sites was accentuated in both CpG islands and single CpGs. We also carried out a pathway analysis using Ingenuity IPA Software. Several canonical pathways appeared differentially methylated. This included, for example, 32 genes of the Wnt-signaling pathway as well as CDK5-signaling genes. Cancer-related genes such as CTNNA2 were also differentially methylated. Taken together, our data provide evidence of a specific PML-RARα associated APL-Methylome. Unexpectedly, primary patient samples exhibited a high degree of relative hypomethylation within published PML-RARα binding sites and across promoter regions in general. Methylation differences were associated with deregulated canonical pathways and especially Wnt-signaling. 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: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 2493-2493
    Abstract: DNA methyltransferases (DNMT) play an important role in regulation of DNA methylation and mutations of DNMT3A are frequently found in AML. In previous studies using a tetracycline-inducible DNMT3B mouse model, we could show that overexpression of DNMT3B affected leukemia initiation and maintenance upon retroviral transduction and serial transplantation of hematopoietic stem and progenitor cells with MSCV-MLL-AF9-GFP and MSCV-cmyc-bcl2-mcherry oncogenic vectors, respectively. Sublethally irradiated recipient mice of DNMT3B overexpressing MLL-AF9 and cmyc/bcl2 leukemic cells developed leukemia with a prolonged latency when compared to recipients of wildtype cells. We performed serial transplantation assays of MLL-AF9 leukemic stem cells, which were sorted for high expression of ckit. The life-prolonging effect of DNMT3B expression was stem cell-specific, as the potential to initiate leukemia was maintained upon serial retransplantation and recipients of DNMT3B overexpressing leukemic stem cells also died significantly later in secondary (p 〈 0.001) and tertiary transplantations (p 〈 0.001). Analysis of global DNA methylation levels in MLL-AF9 ckit+ leukemic stem cells and cmyc/bcl2 leukemic cells via Reduced Representation Bisulfite Sequencing (RRBS) revealed a strong hypermethylation in DNMT3B overexpressing cells, independent of the oncogene used for leukemia induction. Differentially methylated CpG sites were defined as CpGs with at least 20% methylation difference between wildtype and DNMT3B overexpressing samples. Hypermethylation in MLL-AF9 leukemic cells directly correlated with observed hypermethylation in cmyc/bcl2 leukemic cells and inversely correlated with hypomethylation in cmyc/bcl2 cells, indicating that in both leukemias, the same sites are prone to DNMT3B induced DNA methylation. To investigate, if these changes in DNA methylation resulted in different gene expression patterns, we performed microarray analysis of the same MLL-AF9 leukemic wildtype and DNMT3B expressing samples which were also used for DNA methylation analysis. In microarray analyses, we could identify several genes differentially expressed in DNMT3B overexpressing cells when compared to wildtype samples. Interestingly, changes in expression levels could not be attributed to differential DNA methylation in promoter regions. Instead, hypermethylation in exons and gene bodies resulted in downregulation of the respective genes, whereas genes with hypomethylated exons and gene bodies showed higher expression levels. Genes downregulated in DNMT3B overexpressing cells, were mainly cancer-associated genes, which are known to have functions in cellular growth and proliferation, as well as in the hematopoietic system development and maintenance. Gene Set Enrichment Analysis (GSEA) of wildtype cells revealed a strong enrichment of genes upregulated in different stages of hematopoietic stem and progenitor cells as well as in leukemic stem cells, whereas DNMT3B overexpressing samples were enriched in genes which have been shown to be downregulated in hematopoietic and leukemic stem cells and upregulated in mature hematopoietic cells. This strengthens our hypothesis that DNMT3B induced DNA methylation mainly influences the phenotype and function of hematopoietic stem cells and thereby, exerts its inhibitory function on leukemia initiation and maintenance. Taken together, these findings demonstrate that DNMT3B exerts its anti-leukemic effect mainly via induction of aberrant DNA methylation in hematopoietic and leukemic stem cells, thereby changing expression patterns of genes known to be important for stem cell function. The identification of differentially expressed DNMT3B target genes could help to find promising targets for new therapeutic strategies 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: 2013
    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...