Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
Type of Medium
Language
Subjects(RVK)
  • 1
    In: Australian Academic & Research Libraries, Informa UK Limited, Vol. 24, No. 1 ( 1993-01), p. 53-65
    Type of Medium: Online Resource
    ISSN: 0004-8623 , 1839-471X
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 1993
    detail.hit.zdb_id: 2042567-3
    SSG: 24,1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 884-884
    Abstract: In recent studies on acute myeloid leukemia (AML), genes involving DNA methylation (DNMT3A, IDH1/2, TET2) were identified as frequently mutated. Around 17% of AML patients were found to have a mutation in isocitrate dehydrogenase-1 or -2 (IDH1/2). In these patients, 2-hydroxylglutarate (2-HG), which is generated from a neomorphic activity of mutant IDH1/2, accumulates and inhibits DNA hydroxylmethylation mediated by TET family proteins. Interestingly, mutations in the de novo DNA methyltransferase 3A (DNMT3A) and IDH1/2 mutations co–occur in a statistically significant portion of AML patients. In current known mouse models, neither Dnmt3a knockout (KO) and IDH1/2 mutation alone initiates overt hematopoietic malignancy, leading to our hypothesis that Dnmt3a and IDH1/2 mutations may act synergistically to initiate hematopoietic malignancy. To create a Dnmt3a knockout IDH1/2 mutant double mutant mouse model, we have employed a transplantation approach to create Idh2R140Q (one of the abundant IDH2 mutation in AML patients) overexpressing stem cells on a Dnmt3a KO (Dnmt3a–/–) and wild-type (WT) background by retroviral transduction. Idh2WT overexpression was also used as a control group. With a latency of 180 days after transplantation, a hematopoietic disease with a median survival of 197 days was observed in the Dnmt3a–/––Idh2R140Q group. Anemia, thrombocytopenia and monocytosis were observed in morbid Dnmt3a–/––Idh2R140Q mice. The pathological examination of Dnmt3a–/––Idh2R140Q mice showed myelodysplasia in one or more lineages, an accumulation of less differentiated myeloid progenitors in the bone marrow and pronounced extramedullary hematopoiesis in the spleen. Moreover, approximately 20% of Dnmt3a–/––Idh2R140Q mice developed AML. Together, these features led to the diagnosis of MDS/MPN (Myelodysplastic Syndrome / Myeloproliferative Neoplasms) with high transformation rate to AML. In comparison, WT–Idh2R140Q mice also developed less severe MDS/MPN characterized by myeloid differentiation bias and extramedullary hematopoiesis without lethality in one year after bone marrow transplantation. The MDS/MPN of Dnmt3a–/––Idh2R140Q developed into MPD after secondary transplantation with Lin- cKit+ progenitors, while the same progenitors from the control genotypes did not cause hematopoietic diseases in secondary transplantation . The profiling of 2-HG with gas chromatography mass spectrometry (GC-MS) in the serum of morbid mice transplanted with Dnmt3a–/––Idh2R140Q showed an 80-fold increase compared to normal mouse serum, while the 2-HG content in mice transplanted with WT–Idh2R140Q cells was 10-fold higher than that of normal mouse serum. This suggests that Dnmt3a loss-of-function can promote the synthesis of 2-HG by the Idh2R140Q mutation. The metabolomic profiling on cKit+ bone marrow cells identifies 43 metabolites differentially present in malignant Dnmt3a–/––Idh2R140Q cells compared with groups of other genotypes. Furthermore, the unsupervised cluster analysis shows Dnmt3a–/––Idh2R140Q cells have a distinct metabolome profile compared with cells of other genotypes. This suggests a synergistic effect on metabolome between the two genetic backgrounds. The essential amino acid and glycolysis pathway metabolites– are among the most enriched differential present metabolites. In addition, the glutamine anaplerosis pathway is highly upregulated in the Dnmt3a–/––Idh2R140Q group indicated by the increase of a-ketoglutarate and glutamate. In contrast, the WT – Idh2R140Q and Dnmt3a KO groups have no alteration in glutamine anaplerosis pathway metabolites, indicating a synergy on 2-HG synthesis between Dnmt3a knockout and Idh2R140Q. Moreover, an excess of glutamine during in vitro culture significantly promotes the colony forming ability Dnmt3a–/––Idh2R140Q HSPCs, while there’s no effect in Dnmt3 Idh2WT and WT–Idh2R140Q HSPCs. In summary, our research shows for the first time, Dnmt3a loss-of-function promotes 2-HG synthesis with mutant Idh2 and strongly aggravates the phenotype induced by Idh2 mutation. Our research also shows the synergistic effect on the metabolome of two genetic backgrounds. These data likely explain the high frequency of co-mutations between DNMT3A and IDH1/2 that promotes aggressive AML in patients. 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 98, No. 4 ( 2001-08-15), p. 1166-1173
    Abstract: The hematopoietic stem cell underlying acute myeloid leukemia (AML) is controversial. Flow cytometry and the DNA-binding dye Hoechst 33342 were previously used to identify a distinct subset of murine hematopoietic stem cells, termed the side population (SP), which rapidly expels Hoechst dye and can reconstitute the bone marrow of lethally irradiated mice. Here, the prevalence and pathogenic role of SP cells in human AML were investigated. Such cells were found in the bone marrow of more than 80% of 61 patients and had a predominant CD34low/− immunophenotype. Importantly, they carried cytogenetic markers of AML in all 11 cases of active disease examined and in 2 out of 5 cases in complete hematological remission. Comparison of daunorubicin and mitoxantrone fluorescence emission profiles revealed significantly higher drug efflux from leukemic SP cells than from non-SP cells. Three of 28 SP cell transplants generated overt AML-like disease in nonobese diabetic–severe combined immunodeficient mice. Low but persistent numbers of leukemic SP cells were detected by molecular and immunological assays in half of the remaining mice. Taken together, these findings indicate that SP cells are frequently involved in human AML and may be a target for leukemic transformation. They also suggest a mechanism by which SP cells could escape the effects of cytostatic drugs and might eventually contribute to leukemia relapse.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2001
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Society of Hematology ; 2013
    In:  Blood Vol. 122, No. 21 ( 2013-11-15), p. 48-48
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 48-48
    Abstract: The mammalian genome encodes a significant number of long non-coding RNAs (lncRNAs). The functions of some lncRNAs have been determined in biological processes, such as cancer progression, cell-cycle regulation and embryonic stem cell (ESC) pluripotency. However, our understanding of the basic function of lncRNAs in hematopoietic stem cell (HSC) is still limited. Here, we aim to identify the full complement of lncRNAs expressed in mouse HSCs and to determine whether they control HSC function. To uncover lncRNAs expressed in HSC across different ages, we performed RNA-seq on highly purified HSCs (SP-KSL-CD150+) from 4 month (m04), 12 month (m12) and 24 month (m24) old mice. With two biological replicates for each age, deep sequencing generated 368, 311 and 293 million mapped reads for m04, m12 and m24 HSC, respectively. After combining these datasets, assembly of over 1 billion mapped reads for the HSC transcriptome reconstructed 3,104 novel transcripts, which do not correspond to any UCSC, RefSeq or Ensemble known genes. Among them, 2,853 transcripts have multiple assembled exons and a total length 〉 200 bp, representing potential lncRNAs. It has been shown that lncRNAs usually exhibit stage- or cell type-specific expression. To identify lncRNAs specifically expressed in HSC, we further performed RNA-seq on differentiated lymphoid lineage B cells (B220+) and myeloid lineage Granulocytes (Gr1+). Comparison of the expression of these 2,853 transcripts in the three cell types revealed that 173 transcripts are specifically expressed in HSC. As epigenetic mechanisms play critical roles to regulate gene transcription, we checked the chromatin map associated with those novel transcripts by ChIP-seq for H3K4me3, H3K27me3 and H3K36me3 in purified HSCs. Like protein-coding genes, these HSC specific novel transcripts typically contain the H3K4me3 mark at their transcriptional start site (TSS) and H3K36me3 along the gene body. Remarkably, one fifth of those 173 transcripts showed altered expression with HSC aging. Given that HSC function declines with aging, we hypothesize that those transcripts contribute to control HSC homeostasis. We selected three of these transcripts for further validation: LncHSC-1, LncHSC-2 and LncHSC-3. RT-PCR confirmed that they were highly expressed in stem and progenitor populations (KSL), but not in differentiated lineages (B220, CD4, CD8, Mac1, Gr1 and Ter119). Next, we generated retrovirally expressed-miRNA constructs to knockdown these transcripts. In vitro methocult colony forming assay showed that knockdown of LncHSC-1 in HSC significantly increased the colony number after second plating. Lineage analysis revealed that the majority of those cells are c-Kit+, and exhibit similar morphology, possibly representing expanded myeloid progenitors. To confirm our in vitro findings, we further examined their functions in vivo by HSC transplantation. Progenitors in which LncHSC-3 was knocked down failed to contribute to long-term hematopoietic reconstitution, as revealed by loss of retrovirally transduced population in the peripheral blood and bone marrow. In contrast, progenitors in which LncHSC-1 was knocked down resulted in augmented myeloid differentiation, consistent with in vitro CFU results that knockdown increased myeloid colony number. To understand the molecular mechanism through which lncRNAs influence hematopoiesis, we checked gene expression changes upon knockdown of specific transcripts in KSL cells. Overall, 80-100 genes were significantly changed after knockdown of specific transcripts, including cell cycle regulators and chromatin modification enzymes. For example, after LncHSC-3 knockdown, cell cycle regulator Cdkn1a (p21) expression increased, possibly contributing to the inhibition of hematopoietic reconstitution. In summary, here we carried out a comprehensive lncRNAs analysis in HSC and determined HSC specific novel transcripts. Loss-of-function experiments demonstrated that these transcripts may play important roles for HSC self-renewal and differentiation. These findings provide a useful resource to study lncRNA functions in normal hematopoiesis and disease progression. 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Society of Hematology ; 2003
    In:  Blood Vol. 101, No. 6 ( 2003-03-15), p. 2434-2439
    In: Blood, American Society of Hematology, Vol. 101, No. 6 ( 2003-03-15), p. 2434-2439
    Abstract: The CD45 antigen is present on all cells of the hematopoietic lineage. Using a murine model, we have determined whether a lytic CD45 monoclonal antibody can produce persistent aplasia and whether it could facilitate syngeneic or allogeneic stem cell engraftment. After its systemic administration, we found saturating quantities of the antibody on all cells expressing the CD45 antigen, both in marrow and in lymphoid organs. All leukocyte subsets in peripheral blood were markedly diminished during or soon after anti-CD45 treatment, but only the effect on the lymphoid compartment was sustained. In contrast to the prolonged depletion of T and B lymphocytes from the thymus and spleen, peripheral blood neutrophils began to recover within 24 hours after the first anti-CD45 injection and marrow progenitor cells were spared from destruction, despite being coated with saturating quantities of anti-CD45. Given the transient effects of the monoclonal antibody on myelopoiesis and the more persistent effects on lymphopoiesis, we asked whether this agent could contribute to donor hematopoietic engraftment following nonmyeloablative transplantation. Treatment with anti-CD45 alone did not enhance syngeneic engraftment, consistent with its inability to destroy progenitor cells and permit competitive repopulation with syngeneic donor stem cells. By contrast, the combination of anti-CD45 and an otherwise inactive dose of total-body irradiation allowed engraftment of H2 fully allogeneic donor stem cells. We attribute this result to the recipient immunosuppression produced by depletion of CD45+ lymphocytes. Monoclonal antibodies of this type may therefore have an adjunctive role in nonmyeloablative conditioning regimens for allogeneic stem cell transplantation.
    Type of Medium: Online Resource
    ISSN: 1528-0020 , 0006-4971
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2003
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Society of Hematology ; 2012
    In:  Blood Vol. 120, No. 21 ( 2012-11-16), p. 2428-2428
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 2428-2428
    Abstract: Abstract 2428 Aberrant DNA methylation repeatedly has been implicated in cancer development. DNA methyltransferase (DNMT) 3A, which mediates de novo DNA methylation, was found to be mutated in 20% of patients with acute myeloid leukemia and 10% of patients with myelodysplastic syndrome. Recently, mutations associated with myeloid malignancies such as DNMT3A and FLT3 have also been uncovered in patients with early T-cell precursor lymphoblastic leukemia (ETP-ALL) (Neumann et al., 2012; Van Vlierberghe et al., 2011; Zaremba et al., 2012). ETP-ALL is a type of very high-risk ALL associated with myeloid/stem cell gene expression signature and myeloid markers. We have demonstrated that Dnmt3a deletion in mouse causes increased self-renewal of hematopoietic stem cells and an impairment of differentiation (Challen et al., 2011). Dnmt3a loss also produces aberrant methylation associated with oncogenes and tumor suppressor genes. Yet, whether aberrant DNA methylation can drive leukemia remains unknown. As Dnmt3a deletion alone was insufficient for malignancy, secondary mutations are likely necessary for leukemic transformation. Because FLT3 internal tandem duplication (ITD) frequently co-exist with DNMT3A mutations in acute leukemias, we hypothesized that Dnmt3a-loss may cooperate with FLT3-ITD to promote leukemic transformation; and we established a mouse model to test this. Deletion of conditional Dnmt3a with Mx1-cre was induced by injections of pIpC. Subsequently, bone marrow from Dnmt3a-deleted (Dnmt3aKO) donor mice was transduced with MSCV-FLT3-ITD-GFP retrovirus or MSCV-GFP control and transplanted into lethally irradiated recipients. The mice were monitored monthly for development of malignancies by complete blood count and peripheral blood analysis by flow cytometry and followed for disease latency. Moribund mice were sacrificed and analyzed with peripheral blood smears, histology, and immunophenotyping. Dnmt3a deletion with overexpression of FLT3-ITD caused rapid onset T-ALL in 6/8 mice (n=6) with a median latency of 78 days compared to 121 days in WT mice (n=4) overexpressing FLT3-ITD (p & lt;0.0001 Log-rank Mantel-Cox Test) (See figure). Mice from both groups exhibited leukocytosis, splenomegaly, and thymomegaly with high GFP expression detected by FACS. Even after we transduced bone marrow cells enriched for myeloid progenitor and stem cells, Dnmt3a deletion again accelerated T-ALL with median survival of 89 days (n=9) versus 110 days in WT-FLT3-ITD (n=10) mice. T-ALL was observed in 2/4 WT-FLT3-ITD mice and 5/6 Dnmt3aKO-FLT3-ITD mice analyzed (p & lt;0.0001 Log-rank Mantel-Cox Test). By flow cytometry, two distinct types of T-ALL were observed in the bone marrow of Dnmt3a deleted leukemic mice: one was characterized by a double positive population (DP) of CD4+CD8+ lympoblasts (1/6) and another early immature T-cell-like type of CD4-CD8-CD44+CD25-CD11bloCD117+ lymphoblasts (4/6). Gene expression analysis by RT-PCR in the early immature T-ALL showed downregulation of Notch-pathway genes (such as Notch1, Notch 3, Deltex, Hes1) and upregulation of stem cell-associated genes Lyl1 and Scl1, suggesting an ETP-like T-ALL. The ETP-like ALL phenotype has not been seen in WT mice overexpressing FLT3-ITD. The opposite gene expression pattern was seen in the DP population with upregulation of Notch-pathway genes. Furthermore, the DP leukemia was transplantable to secondary recipients within 2 weeks. Whether ETP-like ALL can be transplanted is still under investigation. We are also currently studying the changes in global CpG methylation among the leukemias that have Dnmt3a loss, FLT3-ITD overexpression, and control and also anticipate data from transcriptome analysis by RNA-Seq. These data suggest that stem or progenitor bone marrow cells primed by early loss of Dnmt3a are transformed into DP T-ALL and ETP-like ALL fueled by the overexpression of the oncogene FLT3-ITD. The ETP-like ALL phenotype has not been seen previously in WT mice overexpressing FLT3-ITD, suggesting that Dnmt3a ablation is required. The Dnmt3a-deleted-FLT3-ITD mice with T-ALL is, to our knowledge, the first animal model of human immature T-cell leukemia. This model can enhance our understanding of the pathogenesis of ETP-like ALL with respect to aberrant DNA methylation and will serve as a powerful tool to test novel therapeutic strategies. 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: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 848-848
    Abstract: Abstract 848 DNA methylation is an epigenetic modification in vertebrate genomes critical for regulation of gene expression. DNA methylation is catalyzed by a family of DNA methyltransferase enzymes, Dnmt1, Dnmt3a, and Dnmt3b. Dnmt1 is primarily a maintenance methyltransferase, targeting hemimethylated DNA to reestablish methylation marks after DNA replication. Dnmt3a and Dnmt3b are de novo methyltransferases that are essential for normal embryonic development. In humans, somatic mutations in DNTM3A have been identified in ∼20% of human acute myeloid leukemia (AML) and ∼10% of myelodysplastic syndrome (MDS) patients, but the mechanisms through which these mutations contribute to pathogenesis is not well understood. Congenital mutations in DNMT3B can cause ICF (immunodeficiency, centromeric instability, and facial anomalies) syndrome. These patients exhibit chromosomal instability due to heterochromatin decondensation and demethylation of satellite DNA. Our group has recently reported that Dnmt3a is essential for HSC differentiation (Challen Nature Genetics, 2011). Conditional knockout of Dnmt3a (Dnmt3a-KO) resulted in HSCs that could not sustain peripheral blood generation after serial transplantation, but phenotypically defined HSCs accumulated in the bone marrow. Dnmt3b is also highly expressed in HSCs, but its contribution to gene regulation in hematopoiesis is unclear. Here, we examine the role of Dnmt3b, alone and in combination with Dnmt3a KO, in the regulation of hematopoiesis. We performed conditional ablation of Dnmt3b, as well as Dnmt3a and Dnmt3b simultaneously using the Mx1-cre system. Unlike the Dnmt3a-KO HSCs, loss of Dnmt3b had a minimal impact on blood production. Even after several rounds of transplantation, 3b-KO HSCs performed similarly to WT controls. However, the Dnmt3ab-dKO (double knock-out) peripheral blood contribution was quickly and severely diminished, accompanied by a dramatic accumulation of Dnmt3ab-dKO HSCs in the bone marrow (Figure 1). The dKO phenotype paralleled that of the 3a-KO HSC, but was more extreme. To examine the impact of loss of Dnmt3a and -3b on DNA methylation in HSCs, we performed Whole Genome Bisulfite Sequencing (WGBS) on Dnmt3a-KO, Dnmt3ab- dKO and control HSCs. As we previously found with more limited DNA methylation analysis, loss of Dnmt3a led to both increases and decreases of DNA methylation at distinct genomic regions (Challen, Nature Genetics, 2011). However, loss of both Dnmt3a and -3b primarily resulted in loss of DNA methylation that was much more extensive than that seen in the 3a-KO. In addition, RNAseq of the mutant HSCs revealed increased expression of repetitive elements, inappropriate splicing, and truncation of 3ÕUTRs. To gain insight into the accumulation of Dnmt3ab-dKO HSCs in the bone marrow, we performed a time course analysis of the proliferation and apoptosis status of the HSCs. Every four weeks after transplantation of HSCs, we sacrificed a cohort of 3 control and 3 dKO mice, counted donor derived HSCs in the bone marrow, and analyzed their Ki67 and Annexin V expression. Up to 12 weeks post-transplant, no significant differences are seen in the expression of Ki67 or Annexin V. These data show that while Dnmt3b alone has minimal impact on DNA methylation in HSCs, Dnmt3a and -3b act synergistically to effect gene expression changes that permit HSC differentiation. In the absence of both of these de novo DNA methyltransferases, there is an immediate and extreme shift toward self-renewal of dKO HSCs. The Ki67 and Annexin V expression patterns suggest that a lack of de novo DNA methylation does not affect the proliferation or apoptosis of HSCs, but instead that the accumulation of HSCs and lack of peripheral blood contribution is primarily due to an imbalance between self-renewal and differentiation. By understanding the mechanisms through which Dnmt3a and -3b exert these effects, we should identify genes that are critical for normal hematopoietic differentiation. These genes may serve as targets for therapeutic intervention in malignancies caused by defective DNA methyltransferases. Figure 1: HSC composition of the bone marrow after secondary transplantation of control (left) and double Dnmt3a/3b KO (right) HSCs. After control HSC transplantation, HSCs comprise ∼0.01% of whole bone marrow. After transplantation of dKO HSCs, phenotypically-defined HSCs (KLS CD34–Flk2–) comprise ∼0.48% of bone marrow. Figure 1:. HSC composition of the bone marrow after secondary transplantation of control (left) and double Dnmt3a/3b KO (right) HSCs. After control HSC transplantation, HSCs comprise ∼0.01% of whole bone marrow. After transplantation of dKO HSCs, phenotypically-defined HSCs (KLS CD34–Flk2–) comprise ∼0.48% of bone marrow. 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: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 807-807
    Abstract: Abstract 807 The discovery of JAK2 (JAK2V617F) mutation in polycythemia vera (PV) and essential thrombocythemia (ET) has revolutionized our understanding of PV and ET. However, JAK2V617F is not the disease-initiating mutation and constitutes only part of the clone. Pegylated interferona (PegInfα), a form of interferon-alpha with a longer half-life and more tolerable toxicity, can induce not only hematological remission but also reduction of JAK2V617F allelic burden and suppression of the pre-JAK2V617F PV clone, as demonstrated by the return of polyclonal hematopoiesis (Liu Blood, 03). It is not clear whether this salutary effect is achieved by stimulation of an immune response against the PV clone, preferential activation of normal dormant stem cells (HSCs), or suppression of the PV clone by PegInfα. Using X-chromosome based clonality (qTCA) and JAK2V617F allelic burden quantitative assays, we analyzed selected biomarkers in HSCs, granulocytes and platelets of 18 patients treated with PegInfα, and evaluated cell cycle activity in HSCs. Previous data have indicated that PegInfα modulates the number of CD4+FOXP3+ regulatory T cells (Tregs) in the periphery. This is perhaps an indicator that PegInfα alters immunity against the JAK2V617F-bearing clone. We hypothesized that PegInfα mobilizes marrow Tregs to the periphery, thus decreasing their immunosuppressive and tumor promoting influence on the marrow microenvironment. Therefore, we correlated the number of Tregs before, during and after treatment with JAK2 allelic burden in peripheral blood and marrow samples in ongoing studies. In ongoing studies, 18 patients (7 females and 11 males) with PV and ET were treated with PegInfα, with a median follow-up of 6 months. There were variable JAK2V617F allelic burdens at enrollment (1.2% – 99.8%). Following treatment, 7 of 18 patients demonstrated a decrease in JAK2V617F allelic burden, indicating that PegInfα had an effect on the JAK2V617F-positive clone, while 11 had no response or had an increase in JAK2V617F levels. Six out of 7 female patients were informative for at least one exonic polymorphism out of 5 X-chromosome genes, and all 6 had clonal hematopoiesis as determined by qTCA at the start of treatment. Two of these 6 patients developed polyclonal hematopoiesis during therapy, but in one the JAK2V617F allelic burden was unchanged, indicating that, in this patient, PegInfα had a selective salutary effect on a pre-JAK2V617F clone, while in other patients PegInfα clearly decreased the JAK2V617F-positive clone. In the 7 patients who demonstrated decreased JAK2V617F levels, there was an inverse relationship of JAK2V617F level with the number of circulating peripheral blood Tregs (examples shown in Figs 1 and 2). Statistical analysis of 18 patients showed a significant negative relationship between the number of circulating Tregs and JAK2V617F allelic burden (Spearman's correlation coefficient -0.4308, p 〈 0.001). This suggests that an immunomodulatory effect by PegInfα involving Tregs may, at least in part, be associated with suppression of pre-JAK2V617F and JAK2V617F-positive clonal hematopoiesis. We assessed total colony forming ability in methylcellulose culture media as a measure of hematopoietic progenitor abundance. In 5 patients treated with Pegasys, all 5 demonstrated an increase in colony-forming units. Furthermore, when cell cycle status of CD34+ HSCs was ascertained, we found a decrease in the percentage of HSCs in G0 (quiescence) in 4 out of 6 samples. These findings suggest that PegInfα therapy promotes normal HSC proliferation. For the first time we directly demonstrate that PegInfα therapy in some patients recruits dormant pre-clonal PV HSCs to active hematopoiesis, thereby resulting in production of polyclonal granulocytes and platelets. PegInfα also appears to mobilize Tregs to the circulation, an effect that may be associated with changes in the marrow microenvironment. We conclude that PegInfα can suppress both pre-JAK2V617F and JAK2V617F PV clones in some PV patients, as demonstrated by evaluation of HSCs, granulocytes and platelets, but this often occurs asynchronously. Our preliminary data suggest that PegInfα therapy is associated with an increase in cell cycle activity in HSCs and mobilization of Tregs to the circulation. This provides a possible mechanism by which polyclonal hematopoiesis is restored after PegInfα treatment. 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: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Society of Hematology ; 2012
    In:  Blood Vol. 120, No. 21 ( 2012-11-16), p. 1188-1188
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 1188-1188
    Abstract: Abstract 1188 Chromatin regulators, including both Trithorax-group (trxG) and Polycomb-group (PcG) families, have been reported to maintain hematopoietic stem cells (HSC) self-renewal and differentiation. However, their primary targets in HSC remain to be fully identified. Moreover, those histone modifiers have been recently found to control longevity in C.elegans, which leads us to further investigate their roles in HSC aging. HSCs increase in number, decrease in regeneration capacity, and exhibit a myeloid biased differentiation with age. In this study, we profiled global mRNA (RNA-seq) and chromatin changes (ChIP-seq) in highly purified young (4 month) and old (24 month) murine bone marrow-derived HSCs (SP-KSL-CD150+). One key challenge in this study is that mouse HSCs represent less than 0.01% of all bone marrow cells. Thus, we first developed and optimized a method for successful application of ChIP-seq to a limited number of cells ( 〈 20,000 cells). This method allowed us to generate the binding profiles for H3K4me3, H3K27me3 and H3K36me3 in young and old HSCs, differentiated granulocytes (Gr1+) and B (B220+) cells. In young HSCs, we determined ∼18,000 peaks for the active gene mark H3K4me3. For the repressive mark H3K27me3, we identified ∼6000 peaks across the genome, 2591 of which were present in the promoter region defined as the transcriptional start site (TSS) ± 100 bp. Strikingly, 70.2% (1820 out of 2591) of H3K27me3-enriched genes were also bound by H3K4me3. These so-called “Bivalent genes”, with both H3K4me3 and H3K27me3, are also found in embryonic stem cell (ESC) where they represent master regulators for lineage development. Here we found that these 1820 genes in HSC are prevalent in development, transcriptional regulation and RNA metabolism. They include many lineage-specific transcription factors, such as Cebpa, Ebf-1, Pax5, Gata3, Tbx21, Runx3 and Eomes. During HSC differentiation to granulocytes or B cells, HSC pluripotency regulators acquired the H3K27me3 repressive mark, while lineage regulators lost it in differentiated lineages. In addition, we observed with HSC differentiation alternative promoter usage on many epigenetic modifiers or transcription factors, such as Dnmt3a, Dnmt3b and Kdm6b (Jmjd3) and Runx3. The different isoforms may have different functions in hematopoiesis. Compared to HSC differentiation, HSC aging showed less extensive chromatin changes. Increased H3K4me3 binding at the TSS was identified for ∼300 genes, including Selp, Nupr1, Sdpr, Plscr2, Slamf1 (CD150) and Mt1. Interestingly, several genes in the Hoxb cluster showed increased H3K4me3 binding and higher expression with age. For H3K27me3, although PcG family member EZh2 expression decreases with HSC aging, we did not detect a global H3K27me3 decrease. On the contrary, H3K27me3 binding increased at ∼500 genes and decreased at ∼100 genes. Among them, the lymphoid transcription factors Pax5 and Ebf1 exhibited increased H3K27me3 binding. The enhanced repression of these lymphoid regulators may explain the myeloid-skewed differentiation that occurs with age. We also observed increased H3K27me3 on several Wnt ligands, including Wnt2a, Wnt8a and Wnt8b. As Wnt signaling is required for active HSC cycling, the increased H3K27me3 binding on these ligands may reinforce HSC quiescence. One well known target of the PcG family in aging is Cdkn2a (p16), which showed progressive loss of H3K27me3 repression with aging in neural stem cell (NSC). In contrast, our results revealed that p16 is repressed by H3K27me3 in both 4M and 24M HSC, and we did not observe its expression increase with aging. However, another cell cycle regulator p21 expression increases with aging, accompanied by a decrease of H3K27me3. In summary, we mapped chromatin state alterations with HSC differentiation and aging in this study. These findings will advance our fundamental understanding of HSC biology. Furthermore, chromatin regulators, such as Bmi1 and EZh2, have been implicated in leukemia transformation. This study provides a mechanistic link into how deregulation of these factors correlates with cancer progression. 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: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society of Hematology ; 2007
    In:  Blood Vol. 109, No. 4 ( 2007-02-15), p. 1736-1742
    In: Blood, American Society of Hematology, Vol. 109, No. 4 ( 2007-02-15), p. 1736-1742
    Abstract: A temporal decline in tissue stem cell functionality may be a key component of mammalian aging. The tumor suppressor p53 has recently been implicated as a potential regulator of aging. We examined age-associated hematopoietic stem cell (HSC) dynamics in mice with varying p53 activities. Reduced p53 activity in p53+/− mice was associated with higher numbers of proliferating hematopoietic stem and progenitor cells in old age compared with aged wild-type (p53+/+) mice. We also assessed HSC dynamics in a p53 mutant mouse model (p53+/m) with higher apparent p53 activity than wild-type mice. The p53 hypermorphic (p53+/m) mice display phenotypes of premature aging. Many aged p53+/m organs exhibit reduced cellularity and atrophy, suggesting defects in stem-cell regenerative capacity. HSC numbers from old p53+/m mice fail to increase with age, unlike those of their p53+/+ and p53+/− counterparts. Moreover, transplantation of 500 HSCs from old p53+/m mice into lethally irradiated recipients resulted in reduced engraftment compared with old wild-type p53+/+ and p53+/− HSCs. Thus, alteration of p53 activity affects stem-cell numbers, proliferation potential, and hematopoiesis in older organisms, supporting a model in which aging is caused in part by a decline in tissue stem cell regenerative function.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages