39 research outputs found

    To My Daughter

    Get PDF
    <p>Equal numbers of macaque CD34<sup>+</sup> cells were transduced in 3-d transduction cultures with either the HOXB4GFP or YFP vector and then cultured for an additional 9 d (T02266) or 6 d (K03290 and J02152) in the presence of SCF, TPO, Flt-3L, and G-CSF. All the transduced and expanded cells were infused into myeloablated animals. The percentage of HOXB4GFP<sup>+</sup> and YFP<sup>+</sup> granulocytes was assessed by flow cytometry. Shown is the engraftment of HOXB4GFP<sup>+</sup> and YFP<sup>+</sup> granulocytes after transplantation. (A) T02266, (B) K03290, and (C) J02152. </p

    Venn diagram of genes significantly activated (A) or repressed (B) in Sca1+, Lin- BM cells expressing the ND13, NA10 or ND13(N51S) mutant fusion genes compared to GFP control.

    No full text
    <p>Venn diagram of genes significantly activated (A) or repressed (B) in Sca1+, Lin- BM cells expressing the ND13, NA10 or ND13(N51S) mutant fusion genes compared to GFP control.</p

    Correlation between HOXA7 and HOXA9 (A) and JAG2 and HLF (B) gene expression in human AML analyzed with TaqMan Low Density Array (TLDA).

    No full text
    <p>Spearman rank correlation analysis was done on the log ratio values obtained from the TLDA assay and calculated with the 2<sup>−ΔΔCT</sup> method (n = 34).</p

    Gene expression levels in human AML relative to normal bone marrow.

    No full text
    <p>Gene expression levels in human AML relative to normal bone marrow.</p

    Validation of microarray results with quantitative RT-PCR on unamplified RNA.

    No full text
    <p>Fold changes are calculated against an empty MIG control and <i>Gapdh</i> as endogenous control gene.</p

    Genes changed by NA10 and ND13 but not by the ND13(N51S) mutant compared to the GFP control.

    No full text
    <p>Genes changed by NA10 and ND13 but not by the ND13(N51S) mutant compared to the GFP control.</p

    Meis1 Is Required for Adult Mouse Erythropoiesis, Megakaryopoiesis and Hematopoietic Stem Cell Expansion

    No full text
    <div><p><i>Meis1</i> is recognized as an important transcriptional regulator in hematopoietic development and is strongly implicated in the pathogenesis of leukemia, both as a Hox transcription factor co-factor and independently. Despite the emerging recognition of <i>Meis1</i>’s importance in the context of both normal and leukemic hematopoiesis, there is not yet a full understanding of <i>Meis1</i>’s functions and the relevant pathways and genes mediating its functions. Recently, several conditional mouse models for <i>Meis1</i> have been established. These models highlight a critical role for <i>Meis1</i> in adult mouse hematopoietic stem cells (HSCs) and implicate reactive oxygen species (ROS) as a mediator of <i>Meis1</i> function in this compartment. There are, however, several reported differences between these studies in terms of downstream progenitor populations impacted and effectors of function. In this study, we describe further characterization of a conditional knockout model based on mice carrying a loxP-flanked exon 8 of <i>Meis1</i> which we crossed onto the inducible Cre localization/expression strains, B6;129-<i>Gt(ROSA)26Sor</i><sup><i>tm1(Cre/ERT)Nat</i></sup>/J or B6.Cg-Tg(Mx1-Cre)1Cgn/J. Findings obtained from these two inducible Meis1 knockout models confirm and extend previous reports of the essential role of <i>Meis1</i> in adult HSC maintenance and expansion and provide new evidence that highlights key roles of Meis1 in both megakaryopoiesis and erythropoiesis. Gene expression analyses point to a number of candidate genes involved in Meis1’s role in hematopoiesis. Our data additionally support recent evidence of a role of <i>Meis1</i> in ROS regulation.</p></div

    Loss of <i>Meis1</i> results in an intrinsic defect in LTRC.

    No full text
    <p><b>a)</b> Experimental plan to test the cell-intrinsic requirement for <i>Meis1</i> in HSCs. <b>b)</b> PB engraftment of primary recipients 4 weeks after transplantation and prior to induction shows no difference in engraftment (n = 11). <b>c)</b> PB engraftment of primary recipients 2 days following PolyI:C administration. <i>MxCre</i><sup><i>+</i></sup><i>/Meis1</i><sup><i>-/-</i></sup> (n = 7) engraftment is reduced 17% compared to <i>MxCre/Meis1</i><sup><i>fl/fl</i></sup> (n = 4; <i>p</i> = 0.0005). <b>d)</b> BM engraftment in primary recipients 3 days following PolyI:C administration. <i>MxCre</i><sup><i>+</i></sup><i>/Meis1</i><sup><i>-/-</i></sup> (n = 7) engraftment is reduced 38% compared to <i>MxCre/Meis1</i><sup><i>fl/fl</i></sup> (n = 4; <i>p</i> = 6x10<sup>-6</sup>).</p

    Loss of <i>Meis1</i> results in an intrinsic defect in HSC self-renewal.

    No full text
    <p><b>a)</b> Engraftment of secondary recipients transplanted with bulk CD45.2 cells from primary mice with transplant dose based on the number of CD45.2 cells 24 weeks after transplantation. <b>b)</b> Engraftment of secondary recipients transplanted with sorted CD45.2 cells from primary mice 24 weeks after transplantation.</p

    Gating strategy and representative plots for myeloid progenitors, HSCs and MkP from <i>MxCre/Meis1</i><sup><i>-/-</i></sup> and <i>MxCre/Meis1</i><sup><i>-/+</i></sup> BM

    No full text
    <p>Gating strategy and representative plots for myeloid progenitors, HSCs and MkP from <i>MxCre/Meis1</i><sup><i>-/-</i></sup> and <i>MxCre/Meis1</i><sup><i>-/+</i></sup> BM</p
    corecore