21 research outputs found

    Runx1 exon 6-related alternative splicing isoforms differentially regulate hematopoiesis in mice.

    No full text
    RUNX1 is an important transcription factor for hematopoiesis. There are multiple alternatively spliced isoforms of RUNX1. The best known isoforms are RUNX1a from use of exon 7A and RUNX1b and c from use of exon 7B. RUNX1a has unique functions due to its lack of C-terminal regions common to RUNX1b and c. Here, we report that the ortholog of human RUNX1a was only found in primates. Furthermore, we characterized 3 Runx1 isoforms generated by exon 6 alternative splicing. Runx1bEx6(-) (Runx1b without exon 6) and a unique mouse Runx1bEx6e showed higher colony-forming activity than the full-length Runx1b (Runx1bEx6(+)). They also facilitated the transactivation of Runx1bEx6(+). To gain insight into in vivo functions, we analyzed a knock-in (KI) mouse model that lacks isoforms Runx1b/cEx6(-) and Runx1bEx6e. KI mice had significantly fewer lineage-Sca1(+)c-Kit(+) cells, short-term hematopoietic stem cells (HSCs) and multipotent progenitors than controls. In vivo competitive repopulation assays demonstrated a sevenfold difference of functional HSCs between wild-type and KI mice. Together, our results show that Runx1 isoforms involving exon 6 support high self-renewal capacity in vitro, and their loss results in reduction of the HSC pool in vivo, which underscore the importance of fine-tuning RNA splicing in hematopoiesis

    Cooperation between RUNX1-ETO9a and Novel Transcriptional Partner KLF6 in Upregulation of <i>Alox5</i> in Acute Myeloid Leukemia

    Get PDF
    <div><p>Fusion protein RUNX1-ETO (AML1-ETO, RUNX1-RUNX1T1) is expressed as the result of the 8q22;21q22 translocation [t(8;21)], which is one of the most common chromosomal abnormalities found in acute myeloid leukemia. RUNX1-ETO is thought to promote leukemia development through the aberrant regulation of RUNX1 (AML1) target genes. Repression of these genes occurs via the recruitment of the corepressors N-COR and SMRT due to their interaction with ETO. Mechanisms of RUNX1-ETO target gene upregulation remain less well understood. Here we show that RUNX1-ETO9a, the leukemogenic alternatively spliced transcript expressed from t(8;21), upregulates target gene <i>Alox5</i>, which is a gene critically required for the promotion of chronic myeloid leukemia development by BCR-ABL. Loss of <i>Alox5</i> expression reduces activity of RUNX1-ETO9a, MLL-AF9 and PML-RARα <i>in vitro</i>. However, <i>Alox5</i> is not essential for the induction of leukemia by RUNX1-ETO9a <i>in vivo</i>. Finally, we demonstrate that the upregulation of <i>Alox5</i> by RUNX1-ETO9a occurs via the C<sub>2</sub>H<sub>2</sub> zinc finger transcription factor KLF6, a protein required for early hematopoiesis and yolk sac development. Furthermore, <i>KLF6</i> is specifically upregulated by RUNX1-ETO in human leukemia cells. This identifies KLF6 as a novel mediator of t(8;21) target gene regulation, providing a new mechanism for RUNX1-ETO transcriptional control.</p></div

    <i>Alox5</i> involvement in hematopoietic cell self-renewal.

    No full text
    <p>(A) <i>Alox5</i> required for long-term self-renewal of hematopoietic cells by RE9a. Colony numbers from wildtype or <i>Alox5</i>-/- bone marrow cells transduced with control (MIP) or RE9a retrovirus and serially replated in methylcellulose. Data shown are averages with standard deviations of a representative dataset. Four independent assays were performed. (B) Typical colony images after 9<sup>th</sup> replating from (A) taken using Nikon Eclipse TS100 microscope with 2×/0.06 objective lens and Nikon DS Camera Control Unit DS-U2 system. (C) Flow cytometric analysis of replated cells from (A). Cells from 3<sup>rd</sup>, 6<sup>th</sup> and 9<sup>th</sup> replatings were stained for myeloid lineage markers Gr-1 and CD11b. Representative data from four independent assays shown. (D) and (E) Lack of <i>Alox5</i> decreases colony formation potential of hematopoietic cells transduced with MLL-AF9 and PML-RARα. Wildtype or <i>Alox5</i>-/- bone marrow cells were transduced with MIP and MLL-AF9 (D) or PML-RARα (E) retrovirus and serially replated in methylcellusose. Data shown are averages and standard deviations of representative datasets. Three independent assays were performed.</p

    Loss of <i>Alox5</i> does not block RE9a leukemia induction <i>in vivo</i>.

    No full text
    <p>(A) Survival of mice receiving wildtype or <i>Alox5</i>-/- fetal liver cells transduced by control (MigR1) or RE9a retrovirus. Number of mice in each cohort shown at right. WT median survival: 30.71 weeks; <i>Alox5</i>-/- median survival: 29.43 weeks; <i>p</i> = 0.39. (B) Presence of hematopoietic blast cells in tissues of mice transplanted with RE9a-transduced wildtype or <i>Alox5</i>-/- cells. Peripheral blood smears and cytocentrifugation of bone marrow and spleen cells were stained with Wright-Giemsa solutions. (C) Immunophenotype of myeloid progenitor cells in wildtype and <i>Alox5</i>-/- leukemias. Distribution of EGFP<sup>+</sup>Lin<sup>−</sup>Sca-1<sup>−</sup>c-Kit<sup>+</sup> leukemic cells harvested from spleen shown based on expression of CD34 and Fcγ receptors II/III (FcγRII/III). At least 4 mice analyzed per genotype, with representative distributions shown.</p
    corecore