Setbp1 promotes the self-renewal of murine myeloid progenitors via activation of Hoxa9 and Hoxa10 - PubMed (original) (raw)

. 2012 Jun 21;119(25):6099-108.

doi: 10.1182/blood-2011-10-388710. Epub 2012 May 7.

Yufen Han, Bandana A Vishwakarma, Su Chu, Ravi Bhatia, Kristbjorn O Gudmundsson, Jonathan Keller, Xiongfong Chen, Vasyl Vasko, Nancy A Jenkins, Neal G Copeland, Yang Du

Affiliations

Setbp1 promotes the self-renewal of murine myeloid progenitors via activation of Hoxa9 and Hoxa10

Kevin Oakley et al. Blood. 2012.

Abstract

Acquisition of self-renewal capability by myeloid progenitors to become leukemic stem cells during myeloid leukemia development is poorly understood. Here, we show that Setbp1 overexpression efficiently confers self-renewal capability to myeloid progenitors in vitro, causing their immortalization in the presence of stem cell factor and IL-3. Self-renewal after immortalization requires continuous Setbp1 expression. We also found that Hoxa9 and Hoxa10 mRNA are present at dramatically higher levels in Setbp1-immortalized cells compared with other immortalized cells, and are induced shortly after Setbp1 expression in primary myeloid progenitors. Suppression of either gene in Setbp1-immortalized cells drastically reduces their colony-forming capability. Interestingly, Setbp1 protein associates with Hoxa9 and Hoxa10 promoters in chromatin immunoprecipitation assays in these cells, suggesting that both are direct transcriptional targets of Setbp1. Setbp1 also promotes self-renewal of myeloid progenitors in vivo as its coexpression with BCR/ABL transforms primary mouse myeloid progenitors, generating aggressive leukemias in recipient mice resembling chronic myelogenous leukemia (CML) myeloid blast crisis. Increased SETBP1 mRNA levels were also detected in a subset of CML advanced phase/blast crisis patients with high levels of HOXA9 and HOXA10 expression. Thus, Setbp1 activation represents a novel mechanism conferring self-renewal capability to myeloid progenitors in myeloid leukemia development.

PubMed Disclaimer

Figures

Figure 1

Figure 1

Efficient immortalization of myeloid progenitors by Setbp1 expression. (A) Schematic diagram of the immortalization procedure. (B) Representative cytospin preparation and Wright-Giemsa staining of cells infected with Setbp1 (top panel) or empty retrovirus (bottom panel) at 1 month after infection. Original magnification ×400. Images were obtained using a Nikon Eclipse E800 microscope and a Qimaging Micropublisher 5.0 digital camera. (C) Cytospin preparation of _Setbp1_-immortalized cells (BM70 and P3 population) before and after treatment with G-CSF for 2 days. BM70 cells are immortalized by insertional activation of endogenous Setbp1., Original magnification ×400. (D) Southern blotting analysis of viral integrations present in 6 _Setbp1_-immortalized myeloid progenitor populations (P1-P6) using a _GFP_-specific probe. Seven ug of genomic DNA from each population was digested with _EcoR_I, resulting the generation of a single _GFP_-containing DNA fragment from each provirus. Each band represents an independent integration. (E) Real-time RT-PCR analysis of total RNA isolated from purified LSK, CMP, and GMP populations of C57BL/6 mice using Setbp1_-specific primers. Relative expression levels were calculated by normalizing to β_-actin mRNA levels in the same sample and also in LSK cells. The mean and SD of each relative expression level is shown.

Figure 2

Figure 2

Setbp1 knockdown inhibits the proliferation of _Setbp1_-immortalized myeloid progenitors. (A) Left panel, mean and SD of colony formation potential of BM70 cells in the presence of SCF and IL-3 at 48 hours after infection with a _Setbp1_-specific shRNA (Setbp1-sh) and control shRNA (NC-sh); right panel, representative Western blot analysis of Setbp1 and actin protein in the same infected cells at 72 hours after infection. Relative Setbp1 protein levels after normalization to actin levels in the same sample are indicated. (B) Mean and SD of colony formation potential in the presence of SCF and IL-3 (left panel) and Western blotting analyses (right panel) of S3 cells at 48 and 72 hours, respectively, after infection with _GFP_-specific shRNA (GFP-sh) and control shRNA (NC-sh). (C) Cell cycle distribution of S3 cells infected with _GFP_-specific and control shRNA determined by PI-staining at 96 hours after infection. The mean and SD of each phase is shown (*P < .05).

Figure 3

Figure 3

Setbp1 expression increases Hoxa9 and Hoxa10 mRNA levels. (A) Real-time RT-PCR analysis of total RNA from indicated immortalized progenitor lines using primers specific for Hoxa9 (top panel) and Hoxa10 (bottom panel). Lines immortalized by activation of endogenous Setbp1: SF1 and BM70; lines immortalized by retroviral Setbp1 expression: S1 and S2; _Evi1_-immortalized lines: BM7 and BM83; _Prdm16_-immortalized lines: BM2 and BM36; lines immortalized by unknown mechanisms: BM6 and BM15. Relative expression levels were calculated by normalizing to β-actin mRNA levels in the same sample and also in BM6 cells. The mean and SD of each relative expression level is shown. (B) Real-time RT-PCR analysis of Hoxa9 (top panel) and Hoxa10 (bottom panel) mRNA levels using total RNA from BM progenitors 48 hours after transduction by Setbp1 virus or empty virus. Relative expression levels were calculated by normalizing to β-actin mRNA levels in the same sample and also in cells infected with empty virus (*P < .05).

Figure 4

Figure 4

Hoxa9 and Hoxa10 are essential for the proliferation of _Setbp1_-immortalized myeloid progenitors. (A) Top panel, mean and SD of colony formation potential of BM70 cells in the presence of SCF and IL-3 at 48 hours after infection with _Hoxa9_-specific shRNAs (Hoxa9-sh1 and Hoxa9-sh2) and control shRNA (NC-sh); bottom panel, representative Western blotting analysis of Hoxa9 and actin protein in the infected cells of the top panel at 72 hours after infection. Relative Hoxa9 protein levels after normalization to actin levels in the same sample are indicated. (B) Mean and SD of colony formation potential in the presence of SCF and IL-3 (top panel) and Western blotting analyses (bottom panel) of BM70 cells at 48 and 72 hours, respectively, after infection with _Hoxa10_-specific shRNAs (Hoxa10-sh1 and Hoxa10-sh2) and control shRNA (NC-sh). Relative Hoxa10 protein levels after normalization to actin levels are indicated. (C) Cell cycle distribution of BM70 cells infected with Hoxa9-sh1 and Hoxa10-sh2 and control shRNA determined by PI-staining at 72 hours after infection. The mean and SD of each phase is shown (*P < .05).

Figure 5

Figure 5

Setbp1 directly activates Hoxa9 and Hoxa10 transcription. (A) Amino acid sequence alignments of AT-hook DNA-binding motifs present in human and mouse Setbp1 protein. Amino acid numbers are indicated. The invariable consensus sequence of AT-hook motifs (RGRP) are highlighted in gray. (B-C) ChIP analysis of myeloid progenitors immortalized by the expression of 3xFLAG-tagged Setbp1 using anti-FLAG M2 antibody and control IgG. PCR products using primers specific to various regions of Hoxa9 (B) or Hoxa10 locus (C) were resolved on ethidium bromide-stained agarose gel (top panel). Results are representative of 3 independent experiments. Diagrams of tested region of Hoxa9 and Hoxa10 locus are also shown (bottom panels). Locations of PCR amplicons are indicated as black bars with corresponding numbers. Transcriptional start sites are indicated as arrows. Exons are indicated as white boxes with coding regions highlighted in black.

Figure 6

Figure 6

Setbp1 cooperates with BCR/ABL to transform committed myeloid progenitors. (A) Survival curves of lethally irradiated C57BL/6-Ly5.2 mice receiving 1 × 106 in vitro expanded BM progenitors infected with MSCV-BCR/ABL-IRES-GFP virus, pMYs-Setbp1-IRES-GFP virus, or the combination. Results are combined from 3 independent experiments with each containing the 3 indicated groups. Transduction efficiencies: 28%-42% for BCR/ABL groups; 19%-25% for Setbp1 groups; and 21%-31% for cotransduction groups. Secondary recipients receiving 1 × 106 spleen cells from primary leukemic mice died of leukemia between 11 and 13 days as indicated by the gray bar (n = 5). (B) A typical enlarged leukemic spleen (right) compared with a control normal spleen (left). (C) H&E staining of a typical liver section from leukemic mice displaying liver infiltration by the leukemic cells. Original magnification ×100. (D) Wright-Giemsa staining of cytospin preparation from the BM of a leukemic mouse. Original magnification ×400. (E) Staining of GFP positive leukemic cells from the BM and spleen (SP) of a typical moribund mouse with indicated antibodies. Numbers represent the percentages of gated events. (F) Semiquantitative reverse transcription PCR analysis of total RNA extracted from normal mouse bone marrow (NBM), leukemic BM, and spleen (SP) from 3 leukemic mice (L1, 2, and 3) using indicated gene-specific primers. β-actin was included to control for RNA loading. (G) Southern blotting analysis of genomic DNA from leukemic spleens (L1 to L7) using a _GFP_-specific probe. Samples were digested by _EcoR_I, and each band represents a separate integration. (H) Survival curves of lethally irradiated C57BL/6-Ly5.2 primary recipient mice receiving purified mouse GMPs (1-1.5 × 105 cells/recipient) infected with MSCV-BCR/ABL-IRES-GFP virus, pMYs-Setbp1-IRES-GFP virus, or the combination as well as secondary (2nd), tertiary (3rd), and quaternary (4th) recipients of the cotransduction group receiving 1 × 106 spleen cells from preceding leukemic mice. Transduction efficiencies: 50%-60% for BCR/ABL groups; 40%-45% for Setbp1 groups; and 34%-50% for cotransduction groups.

Figure 7

Figure 7

SETBP1 activation is involved in human CML progression. Real-time RT-PCR analysis of SETBP1, HOXA9, and HOXA10 mRNA levels in total RNA isolated from whole BM of healthy volunteers (normal) and CML chronic phase (CML CP) and advanced/blast crisis phase (CML AP/BC) patients (*samples expressing high SETBP1 mRNA levels). Relative expression levels were calculated by normalizing to BCR mRNA levels in the same sample.

Similar articles

Cited by

References

    1. Dick JE. Acute myeloid leukemia stem cells. Ann N Y Acad Sci. 2005;1044:1–5. - PubMed
    1. Lobo NA, Shimono Y, Qian D, Clarke MF. The biology of cancer stem cells. Annu Rev Cell Dev Biol. 2007;23:675–699. - PubMed
    1. Cozzio A, Passegue E, Ayton PM, Karsunky H, Cleary ML, Weissman IL. Similar MLL-associated leukemias arising from self-renewing stem cells and short-lived myeloid progenitors. Genes Dev. 2003;17(24):3029–3035. - PMC - PubMed
    1. Krivtsov AV, Twomey D, Feng Z, et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature. 2006;442(7104):818–822. - PubMed
    1. Huntly BJ, Shigematsu H, Deguchi K, et al. MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors. Cancer Cell. 2004;6(6):587–596. - PubMed

Publication types

MeSH terms

Substances

LinkOut - more resources