scholarly journals Clinical Response to Traditional Chinese Herbs Containing Realgar (As2S2) is Related to DNA Methylation Patterns in Bone Marrow DNA from Patients with Myelodysplastic Syndrome with Multilineage Dysplasia

2021 ◽  
Vol Volume 13 ◽  
pp. 55-63
Author(s):  
Qing-Bing Zhou ◽  
Yu Du ◽  
Shan-Shan Zhang ◽  
Zheng-Tang Liu ◽  
Rou Ma ◽  
...  
2020 ◽  
pp. jclinpath-2020-207131
Author(s):  
Gabrielle Melo Cavalcante ◽  
Daniela Paula Borges ◽  
Roberta Taiane Germano de Oliveira ◽  
Cristiana Libardi Miranda Furtado ◽  
Ana Paula Negreiros Nunes Alves ◽  
...  

AimsDNA methylation has its distribution influenced by DNA demethylation processes with the catalytic conversion of 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC). Myelodysplastic syndrome (MDS) has been associated with epigenetic dysregulation of genes related to DNA repair system, chronic immune response and cell cycle.MethodsWe evaluated the tissue DNA methylation/hydroxymethylation in bone marrow trephine biopsies of 73 patients with MDS, trying to correlate with the mRNA expression of 21 genes (POLH, POLL, REV3L, POLN, POLQ, POLI, POLK, IRF-1, IRF-2, IRF-3, IRF-4, IRF-5, IRF6, IRF-7, IRF-8,IRF-9, MAD2, CDC20, AURKA, AURKB and TPX2).ResultsThe M-score (5mC) was significantly higher in patients with chromosomal abnormalities than patients with normal karyotype (95% CI –27.127779 to –2.368020; p=0.022). We observed a higher 5mC/5hmC ratio in patients classified as high-risk subtypes compared with low-risk subtypes (95% CI –72.922115 to –1.855662; p=0.040) as well as patients with hypercellular bone marrow compared with patients with normocellular/hypocellular bone marrow (95% CI –69.189259 to –0.511828; p=0.047) and with the presence of dyserythropoiesis (95% CI 17.077703 to 51.331388; p=0.001). DNA pols with translesion activity are significantly influenced by methylation. As 5mC immunoexpression increases, the expressions of POLH (r=−0.816; r2 =0.665; p=0.000), POLQ (r=−0.790; r2=0.624; p=0.001), PCNA (r=−0.635; r2=0.403; p=0.020), POLK (r=−0.633; r2=0.400; p=0.036 and REV1 (r=−0.578; r2=0.334; p=0.049) decrease.ConclusionsOur results confirm that there is an imbalance in the DNA methylation in MDS, influencing the development of chromosomal abnormalities which may be associated with the low expression of DNA polymerases with translesion synthesis polymerases activity.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2693-2693
Author(s):  
Maximilian Mossner ◽  
Olaf Hopfer ◽  
Claudia D Baldus ◽  
Uwe Neumann ◽  
Anke Kmetsch ◽  
...  

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<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.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2781-2781
Author(s):  
Wolf-Karsten Hofmann ◽  
Florian Nolte ◽  
Ouidad Benlasfer ◽  
Eckhard Thiel ◽  
Gerhard Ehninger ◽  
...  

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.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1586-1586
Author(s):  
Christine Richardson ◽  
Rodwell Mabaera ◽  
Christopher H. Lowrey

Abstract Despite years of investigation in a variety of experimental systems, the mechanisms underlying human β-globin locus developmental gene switching remain elusive. Several lines of evidence implicate DNA methylation in this process. As an initial step in studying the role of epigenetic modifications in the human switching process and in determining the mechanisms by which DNA methyltransferase inhibitors reverse the switch, we have characterized the DNA methylation patterns of the individual CpGs in the γ- and β-globin promoters in fetal liver (FL) and adult bone marrow (BM) primary erythroid cells and during in vitro differentiation of adult erythroid cells. Using the bisulfite conversion method we evaluated all CpGs in the ~500 bp regions centered on the γ- and β-globin promoter start sites. Fetal liver (FL) and adult bone marrow (BM) samples were obtained using IRB approved protocols and informed consent procedures. Erythroid cells were purified using anti-glycophorin A (glyA) magnetic beads. Purity was confirmed to be greater than 95%. Samples from five independent BM and FL samples were analyzed and 8–20 bisulfite converted sequences were determined for each promoter in each sample. Our results show that all 8 CpGs between −249 and +210 of the Gγ and Aγ-globin promoters are less than 20% methylated in FL and greater than 80% methylated in BM except for the −158 CpG which is only 40% methylated in BM(p<0.002). The 6 CpGs between −415 and +110 of the β-globin promoter show an inverse pattern with lower levels of DNA methylation in BM. Histone H3 acetylation of the γ-globin promoter, as determined by ChIP analysis, showed a complimentary pattern with higher levels in FL than BM. We next evaluated γ-globin promoter methylation patterns during in vitro erythroid differentiation from CD34+ BM cells. In this experiment, cells were grown with SCF, Flt3 ligand and IL-3 for 7 days and then in EPO for 14 days producing erythroid cells which express 99%HbA and 1% HbF. The initial day 0 CD34+ cells showed 90–100% methylation of all γ promoter sites. By day 3 in culture, before the initiation of erythroid differentiation, methylation at all sites upstream of the promoter had decreased to less than 60% and the CpG at −53 (the site of Stage Selector Protein complex binding) had decreased to less than 20%. The three CpG sites down-stream of the promoter (+6, +17 and +50) remained highly methylated. The pattern was unchanged at day 10, early in erythroid differentiation, when γ-globin mRNA expression was beginning. By day 14, when β-globin expression was peaking, methylation of the upstream promoter had increased back to the 70–100% level at all CpGs. These experiments provide a comprehensive picture of γ- and β-globin promoter methylation during the fetal and adult stages of erythroid development and of the γ-globin promoter during adult erythroid differentiation. The finding of transient γ-promoter hypomethylation during differentiation offers a potential mechanism to explain the transient γ-globin gene expression seen during normal adult erythropoiesis. Our results also raise the possibility that, just as domains of altered histone modification exist in β-globin gene loci, there may also be developmentally-specific domains of DNA methylation.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 608-608
Author(s):  
Matthew J. Walter ◽  
Dong Shen ◽  
Jin Shao ◽  
Li Ding ◽  
Marcus Grillot ◽  
...  

Abstract Abstract 608 Myelodysplastic syndrome (MDS) genomes are characterized by global DNA hypomethylation with concomitant hypermethylation of gene promoter regions compared to CD34+ cells from normal bone marrow samples. Currently, the underlying mechanism of altered DNA methylation in MDS genomes and the critical target genes affected by methylation remain largely unknown. The methylation of CpG dinucleotides in humans is mediated by DNA methyltransferases, including DNMT1, DNMT3A, and DNMT3B. DNMT3A and DNMT3B are the dominant DNA methyltransferases involved in de novo DNA methylation and act independent of replication, whereas DNMT1 acts predominantly during replication to maintain hemimethylated DNA. The function of these proteins in cancer cells is less well defined. Our group recently found that DNMT3A mutations are common in de novo acute myeloid leukemia (62/281 cases, 22%) and are associated with poor survival (Ley, et al, unpublished), providing a rationale for examining the mutation status of DNMT3A in MDS patients. MDS cases (n=150) were classified according to the French-American-British (FAB) system. The patients included refractory anemia (RA; n=67), RA with ringed sideroblasts (RARS; n=5), RA with excess blasts (RAEB; n=72), and RA with excess blasts in transformation (RAEB-T; n=6). The median International Prognostic Scoring System (IPSS) score was 1 (range 0–3), and the median myeloblast count was 4 (range 0–28%). We designed and validated 28 primer pairs covering the coding sequences and splice sites of all 23 exons for DNMT3A. Paired DNA samples were obtained from the bone marrow (tumor) and skin (normal) of each patient so that somatic mutations could be distinguished from inherited variants/polymorphisms. 17,120 reads were produced by capillary sequencing, providing at least 1X coverage for 82.6% of the target sequence (low/no coverage was obtained for 2 out of 28 amplicons). A semiautomated analysis pipeline was used to identify sequence variants and we restricted our analysis to nonsynonymous and splice site nucleotide changes. All mutations were confirmed by independent PCR and sequencing. We identified nonsynonymous DNMT3A mutations in 12/150 bone marrow samples (8% of cases). All the mutations were heterozygous (10 missense, 1 nonsense, 1 frameshift) and were computationally predicted (by SIFT and/or PolyPhen2) to have deleterious functional consequences. DNMT3A mRNA is expressed in normal CD34+ bone marrow cells and was expressed in all MDS patient samples tested (n=28), independent of mutation status. There was no difference in the expression level of total DNMT3A mRNA in CD34+ cells harvested from mutant (n=3) vs. non-mutant MDS samples (n=25). Amino acid R882, located in the methyltransferase domain of DNMT3A, was the most common mutation site, accounting for 4/12 mutations. The clinical characteristics of the 12 patients with DNMT3A mutations were similar to those of the 138 patients without mutations. Specifically, DNMT3A mutations were present in all MDS FAB subtypes (excluding CMML which was not tested) and in patients with IPSS scores ranging from 0–3. Mutations were not associated with a specific karyotype. In addition, there was no correlation between mutation detection and the myeloblast count of the banked bone marrow specimen, suggesting that mutations were not missed due to the cellular heterogeneity in the samples. We compared the overall (OS) and event-free survival (EFS) of the 12 patients with DNMT3A mutations vs. 138 patients without a mutation and observed a significantly worse OS in patients with mutations (p=0.02), with a median survival of 433 and 945 days, respectively. There was a trend towards worse EFS for patients with mutations (p=0.05). A multivariate analysis for outcomes could not be performed due to the small sample size of patients with mutations, indicating that a larger cohort from a clinical trial will be needed to properly address the affect of DNMT3A mutations on outcomes. The small sample size also precluded us from addressing whether the response to the hypomethylating agents 5-azacytidine or decitabine correlated with the mutation status of DNMT3A. If validated in larger cohort studies, we propose that DNMT3A mutation status could help risk stratify de novo MDS patients for more aggressive treatment early in their disease course. Disclosures: Westervelt: Novartis: Honoraria; Celgene: Honoraria, Speakers Bureau. DiPersio:Genzyme: Honoraria.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1194-1194
Author(s):  
Aparna Vasanthakumar ◽  
Hayley Zullow ◽  
Lucy A Godley

Abstract Gender-specific hormones have been known to play a role in hematopoietic function for some time. For example, treatment with estrogens suppresses B lymphocyte production in murine bone marrow, and hormonally compromised mice undergoing hematopoietic stem cell transplantation demonstrate enhanced immune reconstitution. Furthermore, androgens have been employed as therapy for bone marrow failure syndromes. Despite these experimental observations and clinical practices, the precise molecular mechanism by which gender-specific hormones influence physiology is not understood. To test if epigenetic modifications could alter HSC function in a gender-specific manner, we compared the engraftment potential of hematopoietic stem cells (HSCs) with altered DNA methylation patterns in female versus male recipients. We used DNMT3B7 transgenic mice as the HSC source. Our laboratory demonstrated that the introduction of DNMT3B7, a truncated DNMT3B isoform commonly expressed in cancer cells, impedes normal embryonic development. Homozygous DNMT3B7 transgenic mice have developmental defects similar to the Immunodeficiency, Centromeric instability, Facial anomalies syndrome, and demonstrate lymphopenia and defective craniofacial development. These physiological defects are accompanied by global DNA hypermethylation and disruption in DNA methylation patterns (Shah MY et al, Cancer Res. 2010). Since DNMT3B7 homozygous mice fail to survive past the day of birth, we used a transplantation model to assay the effect of DNMT3B7 on hematopoiesis. We found large differences in engraftment potential when cells expressing DNMT3B7 were transplanted into female versus male recipients. Pancytopenia occurred at two weeks, with anemia and leucopenia persisting until eight weeks post-transplantation when females received DNMT3B7 homozygous cells. However, cells from wild-type (WT) embryos engrafted normally regardless of recipient gender. We also observed that oophorectomized female recipients engrafted DNMT3B7-expressing cells normally. Interestingly, we found an improved engraftment of WT cells in these oophorectomized mice, suggesting that female hormones repress hematopoiesis. In competitive transplantation experiments to determine HSC function, the CD45.1 and CD45.2 alleles were used to distinguish competitor and experimental cells respectively. We observed that DNMT3B7-expressing CD45.2+ cells were out-competed by WT CD45.1+ cells within female recipients, although there were 4-fold more transgenic cells than CD45.1+ competitor cells. Because our previous studies suggested that DNMT3B7 functions as a dominant negative isoform of Dnmt3b, we compared our results with DNMT3B7-expressing cells to those observed with competitive transplants using Dnmt3b knockout cells. Cells from WT, heterozygous Dnmt3b, and homozygous Dnmt3b knockout embryos had similar engraftment potentials in female recipients and were not out-competed by competitor WT CD45.1+ cells, similar to previous observations in a distinct Dnmt3b knockout mouse model (Challen GA et al, Nat Genet. 2011). DNMT3B7 homozygous embryos had significantly fewer numbers of HSCs than WT embryos, as assayed by the LSK (Lineage-, Sca1+, Kit+) and SLAM (CD48, CD150) set of markers. We observed a dose-response relative to DNMT3B7 content, with DNMT3B7 homozygous embryos having the fewest number of HSCs, and DNMT3B7 hemizygous embryos having intermediate numbers of HSCs compared to WT embryos. These observations point to the dual influence of epigenetics and hormones on HSC function. Our hope is that we will be able to use our understanding of the molecular basis for the influence of hormonal milieu on hematopoiesis to augment stem/progenitor cell function in patients undergoing stem cell transplantation and chemotherapy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 734-734
Author(s):  
Grant A Challen ◽  
Allison Mayle ◽  
Deqiang Sun ◽  
Mira Jeong ◽  
Min Luo ◽  
...  

Abstract DNA methylation is one of the major epigenetic modifications in the vertebrate genome and is important for development, stem cell differentiation, and malignant transformation. DNA methylation is catalyzed by the DNA methyltransferase enzymes Dnmt1, Dnmt3a, and Dnmt3b. We have recently shown that Dnmt3a is essential for hematopoietic stem cell (HSC) differentiation. Ablation of Dnmt3a in hematopoietic cells (Mx1-CRE; Dnmt3a-KO) resulted in HSCs that could not sustain peripheral blood generation after serial transplantation, while phenotypically defined HSCs accumulated in the bone marrow. Recurrent somatic mutations in DNTM3A have been discovered in patients with a wide range of hematopoietic malignancies (AML, MDS, MPN, CML, T-ALL, T-cell lymphoma) suggesting a critical role for de novo DNA methylation in normal and leukemic hematopoiesis. As Dnmt3b is also highly expressed in HSCs and congenital mutations in DNMT3B can cause ICF (immunodeficiency, centromeric instability, and facial anomalies) syndrome, in this study we used a mouse model to investigate if Dnmt3b had distinct roles in HSCs. We conditionally inactivated Dnmt3b in HSCs using the Mx1-CRE system (Dnmt3b-KO) and performed serial competitive transplantation. Loss of Dnmt3b had minimal functional consequences for adult HSC function even after three rounds of transplantation. However, combinatorial deletion of both Dnmt3a and Dnmt3b (Dnmt3ab-dKO) exacerbated the differentiation defect seen in Dnmt3a-KO HSCs, leading to a dramatic accumulation of mutant HSCs in the bone marrow (>50-fold), suggesting a synergistic effect resulting from simultaneous ablation of both de novo DNA methyltransferases. The accumulation of Dnmt3ab-dKO HSCs cannot be attributed to altered proliferation or apoptosis, but is due to an imbalance between self-renewal and differentiation. RNA-SEQ of the mutant HSCs revealed loss of transcriptional integrity in Dnmt3ab-dKO HSCs including increased expression of repetitive elements, inappropriate mRNA splicing, and over-expression of HSC-specific genes. To examine the impact of loss of Dnmt3a and -3b on DNA methylation in HSCs, we performed Whole Genome Bisulfite Sequencing (WGBS). Ablation of both enzymes resulted in loss of DNA methylation that was much more extensive than that seen in the absence of Dnmt3a alone, while loss of Dnmt3b alone showed only minimal changes in DNA methylation compared to control HSCs. One puzzling finding was the observation that a subset of promoter CpG islands (CGIs) actually gained DNA methylation in Dnmt3a-KO HSCs. This CGI hypermethylation is a cancer methylome phenotype and was specific to Dnmt3a-KO HSCs (Figure 1A). The HSC transplant experiments suggest that Dnmt3a can compensate for Dnmt3b in HSCs, but Dnmt3b cannot reciprocate in the reverse situation. An explanation for increases in DNA methylation is that in the absence of Dnmt3a, abnormal function of Dnmt3b may lead to aberrant CGI hypermethylation as the hypermethylation was lost when both enzymes were conditionally inactivated. To confirm the mechanism, post-transplant Dnmt3ab-dKO HSCs were transduced with a retroviral vector encoding ectopic expression of Dnmt3b (MIG-Dnmt3b) or a control empty vector (MIG) and assessed for DNA methylation by bisulfite PCR. Using the promoter CGI of Praf2 as an example, enforced expression of Dnmt3b in Dnmt3ab-dKO HSCs resulted in increased DNA methylation at this loci compared to Dnmt3ab-dKO HSCs transduced with a control empty vector (MIG), control HSCs transduced with either MIG or MIG-Dnmt3b and untransduced HSCs (Figure 1B). It is possible that when Dnmt3b tries to compensate for Dnmt3a, the locus-specificity for targets is reduced, leading to aberrant DNA methylation patterns. Promoter CGI hypermethylation is a cancer phenotype observed in a wide range of tumors, including hematopoietic neoplasms driven by mutation in DNMT3A. Targeting DNMT3B in DNMT3A-mutation hematopoietic pathologies may be a therapeutic option for restoring normal DNA methylation and gene expression patterns.Figure 1Praf2 promoter DNA methylation. Open circle = unmethylated CpG, closed circle = methylated CpG. (A) DNA methylation patterns in control (Ctl), Dnmt3a-KO (3aKO), Dnmt3b-KO (3bKO) and Dnmt3ab-dKO HSCs (dKO). (B) Patterns in control and Dnmt3ab-dKO HSCs transduced with empty vector (MIG) or ectopic Dnmt3b, compared to untransduced HSCs.Figure 1. Praf2 promoter DNA methylation. Open circle = unmethylated CpG, closed circle = methylated CpG. (A) DNA methylation patterns in control (Ctl), Dnmt3a-KO (3aKO), Dnmt3b-KO (3bKO) and Dnmt3ab-dKO HSCs (dKO). (B) Patterns in control and Dnmt3ab-dKO HSCs transduced with empty vector (MIG) or ectopic Dnmt3b, compared to untransduced HSCs. Disclosures: No relevant conflicts of interest to declare.


2012 ◽  
Vol 2012 ◽  
pp. 1-4
Author(s):  
Michael Gilbertson ◽  
Annabel Tuckfield ◽  
Surender Juneja

We present the case of a 70-year-old woman who had a bone marrow examination performed to investigate marked thrombocytopenia in the context of a recent history of metastatic glucagonoma. Surprisingly this identified marked dysmegakaryopoiesis and fulfilled diagnostic criteria for refractory cytopenia with multilineage dysplasia, with a relatively uncommon associated cytogenetic lesion t(1;7). We present the case and review the literature of this cytogenetic lesion.


Author(s):  
Anna Hecht ◽  
Julia A. Meyer ◽  
Johann-Christoph Jann ◽  
Katja Sockel ◽  
Aristoteles Giagounidis ◽  
...  

AbstractMyelodysplastic syndrome (MDS) with isolated deletion of chromosome 5q (MDS del5q) is a distinct subtype of MDS with quite favorable prognosis and excellent response to treatment with lenalidomide. Still, a relevant percentage of patients do not respond to lenalidomide and even experience progression to acute myeloid leukemia (AML). In this study, we aimed to investigate whether global DNA methylation patterns could predict response to lenalidomide. Genome-wide DNA methylation analysis using Illumina 450k methylation arrays was performed on n=51 patients with MDS del5q who were uniformly treated with lenalidomide in a prospective multicenter trial of the German MDS study group. To study potential direct effects of lenalidomide on DNA methylation, 17 paired samples pre- and post-treatment were analyzed. Our results revealed no relevant effect of lenalidomide on methylation status. Furthermore, methylation patterns prior to therapy could not predict lenalidomide response. However, methylation clustering identified a group of patients with a trend towards inferior overall survival. These patients showed hypermethylation of several interesting target genes, including genes of relevant signaling pathways, potentially indicating the evaluation of novel therapeutic targets.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 391-391
Author(s):  
Amber Hogart ◽  
Subramanian S. Ajay ◽  
Hatice Ozel Abaan ◽  
Stacie M. Anderson ◽  
Elliott H. Margulies ◽  
...  

Abstract Abstract 391 DNA methylation is a reversible epigenetic modification that is required for proper mammalian development and is proposed to contribute to the pathogenesis of hematologic diseases including leukemia and bone marrow failure syndromes. Elucidating the pathways and genes regulated by DNA methylation during hematopoiesis may reveal new therapeutic targets for disease. Because the phenotype and activity of hematopoietic stem cells (HSC) and hematopoietic progenitor cells of many different lineages have been defined by both in vitro and in vivo assays, hematopoiesis is an excellent model for investigating epigenomic changes during differentiation. HSCs have the ability to self-renew and to generate blood cells of all lineages, which allows them to repopulate recipients after stem cell transplantation. The common myeloid progenitor (CMP) gives rise to all myeloid cell types including neutrophils, monocytes, platelets, and red blood cells, but cannot self renew or repopulate. In contrast to the multipotent HSC and CMP, erythroblasts (ERY) are terminally committed cells that become mature enucleated red blood cells. These three cell types represent unique stages of lineage commitment with distinct transcriptional programs, and potentially unique epigenomic signatures. In contrast to human HSC, which are defined by the absence of several cell surface markers, mouse HSC have the cell surface phenotype of lineage marker negative (Lin-) c-kit+ Sca-1+ and can be positively selected. For this reason we chose the mouse model for genome-wide methylation profiling. Murine HSC and CMP (Lin- c-kit+ Sca-1-) cells were enriched from adult mouse bone marrow with flow cytometry. Erythroblasts (CD71+/Ter119+) were positively selected from E13.5 mouse fetal livers. Genomic DNA isolated from each enriched cell population was sheared to 200-300 bp fragments. MBD2, one of five endogenous mammalian methyl CpG binding domain proteins, binds methylated DNA sequences with broad affinity. Methylated DNA fragments were enriched from the genomic DNA using a tagged, recombinant MBD2 pulldown kit (Active Motif). After pulldown, enrichment of known methylated sequences regulating the imprints of Snrpn and Rasgrf was validated by qPCR. Two biological replicates of HSC, CMP, and ERY methylated sequences and negative control supernatant fractions were submitted for high-throughput sequencing with the Illumina Genome Analyzer platform. Raw sequence data containing 32-46 × 106 reads of 36-50 base pairs were obtained for each sample. The Eland program was used to map 41-59% of reads to unique sequences in the mouse genome. Model-based Analysis of ChIP-Seq (MACS) was used to estimate the mean and variance of the sequence tag distribution across the genome and define peaks below the significance threshold of p<10-5. The number of methylation peaks decreased as cells differentiated, with 64,000 peaks identified in HSC (24,000 unique), 41,000 peaks in CMP (2000 unique), and 23,000 peaks in ERY (1000 unique). Approximately 20,000 peaks were common between all cell types with 57% of these peaks residing in RefSeq genes, 8% in regions adjacent to RefSeq genes (<10 kb), and 35% of methylation peaks in intergenic regions. Comparison of HSC expression data from Akashi et al (Blood 101: 383, 2003) to our HSC genic methylation peaks revealed that 2/3 of HSC genic peaks are within transcriptionally silent genes while 1/3 of HSC genic peaks are within expressed genes. Although DNA methylation is often associated with gene silencing, the important developmental gene Gata2 contains methylation peaks in HSC and CMP, cells that express Gata2, that are absent in ERY, where Gata2 is repressed. A Gata1-Fog1-Mbd2 complex has been described by Rodriguez et al (EMBO 24: 2354, 2005), therefore providing a link between DNA methylation and proteins known to bind at the Gata2 locus. Grass et al (Mol. Cell. Biol. 26:7056, 2006) determined that Gata2 is regulated by long-range interactions of GATA protein complexes, and consistent with this observation, distinct methylation patterns are observed up to 100 kb upstream of the Gata2 gene. Our genome-wide analysis supports an association of methylation with gene silencing but also suggests that DNA methylation is a dynamic epigenetic mark that influences hematopoietic differentiation. The changes in DNA methylation we observe around Gata2 may also contribute to long-range chromatin organization. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document