Dnmt3b Has Few Specific Functions In Adult Hematopoietic Stem Cells But Shows Abnormal Activity In The Absence Of Dnmt3a

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.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1251-1251
Author(s):  
Kasper D Rasmussen ◽  
Guangshuai Jia ◽  
Jens V. Johansen ◽  
Olivier A. Bernard ◽  
Kristian Helin

Abstract Disruption of the epigenome can contribute to both cancer initiation and tumor progression. The methylcytosine dioxygenase TET2, which is involved in the regulation DNA methylation patterns, is frequently mutated in Acute Myeloid Leukemia (AML) patients and plays a central role in normal hematopoiesis. However, Tet2–deficient mouse models do not readily develop acute leukemia suggesting that additional oncogenic events are necessary for full-blown transformation. The translocation t(8;21)(q22;q22) results in expression of the AML1-ETO (AE) fusion protein and is present in ∼5-10% of AML patients. Interestingly, the combination of t(8;21)(q22;q22) and TET2 mutations, although rare, have been observed in several studies in patients groups covering both pediatric and adult de novo AML. To confirm a direct role of TET2 mutations in the development of t(8;21)(q22;q22)-rearranged leukemias we expressed the AE fusion protein in hematopoietic stem and progenitor cells (HSPC) isolated from WT or Tet2-deficient animals. Serial replating assays in semisolid media as well as growth in liquid culture suggested a cooperative effect of AE and Tet2 deficiency as indicated by a significant increase in proliferation and colony formation efficiency. We then used a bone marrow transplantation model to assess the in vivo leukemic potential of AE;Tet2-/- HSPCs. The majority of recipient mice transplanted with AE;Tet2-/- (n=5/6) HSPCs succumbed to an aggressive and transplantable AML-like condition with a median latency of 5-6 months, whereas mice receiving AE HSPCs remained healthy over a 12 month observation period (n=6/6). The morbid mice showed many signs of disease including loss of weight, anemia, splenomegaly and large number of leukemic blasts (CD45neg) in circulation. Further analysis of the bone marrow and spleen revealed a massive expansion of a linneg (CD3e, Gr-1, Mac-1, B220, Ter119), c-Kitlow-high, CD16/32pos cell population indicating a block of differentiation and expansion of an immature myeloid progenitor population from AE:Tet2-/- HSPCs. To investigate how Tet2-deficiency contributes to transformation of t(8;21)(q22;q22)-rearranged cells we generated and cultured pre-leukemic AE-transduced cells in vitro from Tet2fl/fl HPSCs harboring the inducible Rosa26-CreERT2 allele. Genome-wide analysis of methylation patterns and gene expression showed that induction of Tet2-deficiency in pre-leukemic cells leads to progressive hypermethylation of gene-regulatory elements and altered expression of several genes implicated in tumorigenesis. Thus, this study illustrates how aberrant DNA methylation patterns can contribute to disease and confirm the role of Tet2 as a tumor suppressor preventing leukemogenesis. Disclosures: Helin: EpiTherapeutics : Consultancy, Equity Ownership.


2010 ◽  
Vol 21 (12) ◽  
pp. 2066-2077 ◽  
Author(s):  
Anita L. Sørensen ◽  
Bente Marie Jacobsen ◽  
Andrew H. Reiner ◽  
Ingrid S. Andersen ◽  
Philippe Collas

Mesenchymal stem cells (MSCs) isolated from various tissues share common phenotypic and functional properties. However, intrinsic molecular evidence supporting these observations has been lacking. Here, we unravel overlapping genome-wide promoter DNA methylation patterns between MSCs from adipose tissue, bone marrow, and skeletal muscle, whereas hematopoietic progenitors are more epigenetically distant from MSCs as a whole. Commonly hypermethylated genes are enriched in signaling, metabolic, and developmental functions, whereas genes hypermethylated only in MSCs are associated with early development functions. We find that most lineage-specification promoters are DNA hypomethylated and harbor a combination of trimethylated H3K4 and H3K27, whereas early developmental genes are DNA hypermethylated with or without H3K27 methylation. Promoter DNA methylation patterns of differentiated cells are largely established at the progenitor stage; yet, differentiation segregates a minor fraction of the commonly hypermethylated promoters, generating greater epigenetic divergence between differentiated cell types than between their undifferentiated counterparts. We also show an effect of promoter CpG content on methylation dynamics upon differentiation and distinct methylation profiles on transcriptionally active and inactive promoters. We infer that methylation state of lineage-specific promoters in MSCs is not a primary determinant of differentiation capacity. Our results support the view of a common origin of mesenchymal progenitors.


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.


2010 ◽  
Vol 19 (8) ◽  
pp. 1257-1266 ◽  
Author(s):  
Anita L. Sørensen ◽  
Sanna Timoskainen ◽  
Franklin D. West ◽  
Kristin Vekterud ◽  
Andrew C. Boquest ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 835-835 ◽  
Author(s):  
Grant A. Challen ◽  
Jonathan S Berg ◽  
Margaret A. Goodell

Abstract Abstract 835 DNA methylation is one of the major epigenetic modifications in the vertebrate genome and is catalyzed by the DNA methyltransferase enzymes Dnmt1, Dnmt3a and Dnmt3b. We observed a dynamic expression profile of Dnmt3a and Dnmt3b in the hematopoietic system with both enzymes expressed at exponentially higher levels in hematopoietic stem cells (HSCs) compared to progenitors and differentiated cells and hypothesized that some of the unique characteristics of HSCs were epigenetically regulated by Dnmt3a and Dnmt3b. To study this, we crossed Dnmt3a and -3b conditional knock-out (KO) mice to Mx1-cre mice to generate inducible single- and double-KO (dKO) mice. We performed competitive transplantation of HSCs (side-population+c-Kit+Lineage-Sca-1+ = SPKLS) from these mice along with wild-type whole bone marrow competitor and induced deletion of the Dnmt3's in the donor cells by sequential pIpC injections in the wild-type recipients. No dramatic differences were observed in primary recipients in the absence of other hematopoietic perturbation, however when we re-transplanted Dnmt3a- and Dnmt3b-KO HSCs into secondary recipients, they exhibited surprisingly high peripheral blood reconstitution compared to control HSCs (>4-fold increase in engraftment). This was reflected in the bone marrow of these mice with a corresponding >4-fold expansion of the HSC pool (phenotypically defined by any of SPKLS; CD34-Flk2-KLS; CD150+CD48-KLS) with virtually all of these cells being derived from the Dnmt3a- and Dnmt3b-KO donor HSCs. Consistent with a previous study, we observed a decline in functional output of Dnmt3a/3b-dKO HSCs in secondary transplants in terms of peripheral blood chimerism, but surprisingly these mice also exhibited a modest expansion of the HSC pool (∼2-fold), the majority of which were derived from donor Dnmt3a/3b-dKO HSCs. In subsequent tertiary and quaternary transplantation, Dnmt3 single-KO HSCs remained highly superior in peripheral blood engraftment capacity relative to control HSCs and Dnmt3a/3b-dKO HSCs (Figure 1), although the expansion of the HSC pool in all Dnmt3-KOs continued to varying degrees. This enhanced HSC activity appears to be a cell autonomous mechanism as purified Dnmt3-KO SPKLS cells from transplanted mice have much greater hematopoietic colony forming potential in vitro compared to control HSCs on a per cell basis. However the observed HSC expansion does not appear attributable to either enhanced proliferation of Dnmt3-KO HSCs or more resistance to apoptosis. The serially-transplanted Dnmt3-KO HSCs are not overtly transformed, in that the levels of differentiated blood cells are still normal and the mice appear to be healthy. This may be akin to a pre-malignant state seen in human myelodysplastic syndrome. We have performed microarray expression profiling of serially-transplanted Dnmt3-KO HSCs and identified several candidate genes which are currently being investigated as the mechanism for HSC expansion. Our data suggest ablation of de novo DNA methylation in HSCs uncouples normal self-renewal and differentiation. These studies present further evidence for the contribution of epigenetic regulation to stem cell activity and provide a tantalizing link between potential aberrant methylation in HSCs contributing to leukemic transformation. Disclosures: No relevant conflicts of interest to declare.


2010 ◽  
Vol 104 (7) ◽  
pp. 935-940 ◽  
Author(s):  
Alexandra Fischer ◽  
Sonja Gaedicke ◽  
Jan Frank ◽  
Frank Döring ◽  
Gerald Rimbach

The aim of the present study was to determine the effects of a 6-month dietary vitamin E (VE) deficiency on DNA methylation and gene expression in rat liver. Two enzymes, 5-α-steroid reductase type 1 (SRD5A1) and the regulatory subunit of γ-glutamylcysteinyl synthetase (GCLM), which are differentially expressed on the mRNA level, were analysed for promoter methylation in putative cytosine-phospho-guanine (CpG) island regions located at the 5′ end using base-specific cleavage and matrix-assisted laser desorption ionisation time-of-flight MS. A twofold increase in the mRNA level of SRD5A1 gene and a twofold decrease in the mRNA level of GCLM gene in VE-deficient animals were not associated with different CpG methylation of the analysed promoter region. Furthermore, global DNA methylation was not significantly different in these two groups. Thus, the present results indicate that the VE-induced regulation of SRD5A1 and GCLM in rat liver is not directly mediated by changes in promoter DNA methylation.


2018 ◽  
Author(s):  
Tanja Božić ◽  
Joana Frobel ◽  
Annamarija Raic ◽  
Fabio Ticconi ◽  
Chao-Chung Kuo ◽  
...  

AbstractThede novoDNA methyltransferase 3A (DNMT3A) plays pivotal roles in hematopoietic differentiation. In this study, we followed the hypothesis that alternative splicing ofDNMT3Ahas characteristic epigenetic and functional sequels. SpecificDNMT3Atranscripts were either downregulated or overexpressed in human hematopoietic stem and progenitor cells and this resulted in complementary and transcript-specific DNA methylation and gene expression changes. Functional analysis indicated that particularly transcript 2 (coding for DNMT3A2) activates proliferation and induces loss of a primitive immunophenotype, whereas transcript 4 interferes with colony formation of the erythroid lineage. Notably, in acute myeloid leukemia (AML) expression of transcript 2 correlates with itsin vitroDNA methylation and gene expression signatures and is associated with overall survival, indicating thatDNMT3Avariants impact also on malignancies. Our results demonstrate that specificDNMT3Avariants have distinct epigenetic and functional impact. Particularly DNMT3A2 triggers hematopoietic differentiation and the corresponding signatures are reflected in AML.


Sign in / Sign up

Export Citation Format

Share Document