scholarly journals Gfi-1 regulates the erythroid transcription factor network through Id2 repression in murine hematopoietic progenitor cells

Blood ◽  
2014 ◽  
Vol 124 (10) ◽  
pp. 1586-1596 ◽  
Author(s):  
Wonil Kim ◽  
Kimberly D. Klarmann ◽  
Jonathan R. Keller

Key Points Reducing Id2 in Gfi-1−/− mice restores radioprotective function of hematopoietic progenitors and partially rescues erythroid development. Rescue of erythroid development in Gfi-1−/− mice by Id2 reduction directly correlates with an increase of Gata1, Eklf, and EpoR expression.

2014 ◽  
Vol 194 (3) ◽  
pp. 940-949 ◽  
Author(s):  
Michelle L. Ratliff ◽  
Julie M. Ward ◽  
Joan T. Merrill ◽  
Judith A. James ◽  
Carol F. Webb

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1425-1425
Author(s):  
Alla Dolnikov ◽  
Ailyn Choo ◽  
Patricia Palladinetti ◽  
Toby Passioura ◽  
Geoff Symonds ◽  
...  

Abstract Activating mutations of the Ras genes occur at high frequency in acute myeloid leukemia (AML). We have previously shown that expression of mutant N-ras(N-rasm) in murine hematopoietic stem cells is sufficient to induce a myeloid malignancy that resembles human AML(Mackenzie et al. Blood, 1999, 93, 2043–2056). In a ’humanised’ NOD/SCID mouse model N-rasm induced a pre-leukemic condition characterised by myeloid proliferation of human hematopoietic progenitor cells in the bone marrow of recipient mice (Shen et al. Exp. Hematol., 2004, 32: 852–860). Even though Ras usually acts as a dominant transforming oncogene, in primary cells and some cancer cell lines, Ras inhibits cell growth. We have previously shown that ectopic expression of N-rasm in leukemia U937 and K562 cells leads to growth suppression (Passioura et al. Cancer Res. 2005, 65, 797–804). The expression profile induced by N-rasm in these cells included the up-regulation of transcription factor Interferon Regulatory Factor1 (IRF1) and activation of cdk inhibitor p21WAF. IRF1 was previously defined as a tumour suppressor, and as such is a target of oncogenic mutations in AML. Antisense suppression of IRF1 prevented N-rasm induced growth suppression and up-regulation of p21WAF1. These results defined a novel tumour suppressive response to oncogenic N-rasm in leukemia cells. A retroviral cDNA library screen for genes that counteract N-rasm-induced growth suppression identified the gene for the Interferon Regulatory Factor2 (IRF2), and as confirmation of the screen, over-expression of IRF2 in leukemia U937 cells acted to inhibit N-rasm-induced growth suppression (Passioura et al. Oncogene. 2005; 24: 7327–36). IRF2 is known for its oncogenic properties and can antagonise IRF1-mediated tumour suppression. In addition, IRF2 is often up-regulated in primary leukemia samples. Here we show that IRF2 gene suppression using RNA interference acts to suppress the growth of leukemia TF-1 cells bearing N-ras mutation in codon 61 and expressing high levels of IRF1 and IRF2 and low level of p21Waf1. IRF2 down-regulation confirmed at RNA (quantitative RT-PCR) and protein (Western analysis) levels resulted in up-regulation of p21Waf1 and G2/M- rather than G1/S-growth arrest. In addition, increased polyploidisation that results from discoordinated DNA synthesis in mitotically arrested cells, was observed. In addition, IRF2-down-regulation significantly reduced clonogenic growth of the leukemic blasts. Cell growth of normal hematopoietic progenitor cells that express low levels of both IRF1 and IRF2, however, was not affected by IRF2 targeting. IRF2 targeting is currently being examined in primary AML samples in an animal model of AML. We suggest that IRF2 suppression can be used for ex vivo purging of leukemia cells in the autologous stem cell transplantation setting. To the best of our knowledge, specific IRF2 inhibition in cancer cells as a potential therapeutic approach has not been tested to date. IRF2 suppression may prove to be a novel therapeutic target for leukemia therapy.


Blood ◽  
1998 ◽  
Vol 91 (8) ◽  
pp. 2672-2678 ◽  
Author(s):  
Morgan Jenkins ◽  
Mary Beth Hanley ◽  
Mary Beth Moreno ◽  
Eric Wieder ◽  
Joseph M. McCune

It is still uncertain whether multilineage hematopoietic progenitor cells are affected by human immunodeficiency virus-1 (HIV-1) infection in vivo. The SCID-hu Thy/Liv model is permissive of long-term multilineage human hematopoiesis, including T lymphopoiesis. This model was used to investigate the effects of HIV-1 infection on early hematopoietic progenitor function. We found that both lineage-restricted and multilineage hematopoietic progenitors were depleted from grafts infected with either a molecular clone or a primary isolate of HIV-1. Depletion of hematopoietic progenitors (including CD34+ cells, colony-forming units in methylcellulose, and long-term culture-initiating cells) occurred several days before the onset of thymocyte depletion, indicating that the subsequent rapid decline in thymocyte numbers was due at least in part to loss of thymocyte progenitors. HIV-1 proviral genomes were not detected at high frequency in hematopoietic cells earlier than the intrathymic T-progenitor cell stage, despite the depletion of such cells in infected grafts. Proviral genomes were also not detected in colonies derived from progenitor cells from infected grafts. These data demonstrate that HIV-1 infection interrupts both lineage-restricted and multilineage hematopoiesis in vivo and suggest that depletion of early hematopoietic progenitor cells occurs in the absence of direct viral infection.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 509-509 ◽  
Author(s):  
Safak Yalcin ◽  
Sathish Kumar Mungamuri ◽  
Dragan Marinkovic ◽  
Xin Zhang ◽  
Wei Tong ◽  
...  

Abstract Reactive oxygen species (ROS) are toxic byproducts of oxidative metabolism implicated in many debilitating human disorders including hematological malignancies and aging. ROS are also generated by growth factors and cytokine stimulation and play critical functions in normal cellular signaling. However, not much is known of how ROS impact physiological processes in normal and diseased states. We and others have recently shown critical functions for box (O) family of forkhead transcription factors (Fox)O in the regulation of physiological ROS in primitive hematopoietic cells. In particular, FoxO3 has emerged as the principal FoxO whose regulation of ROS is essential for the maintenance of hematopoietic stem cell pool. Although FoxO3’s activity is constitutively repressed by several oncoproteins that play critical roles in myeloproliferative disorders the role of FoxO3 in the regulation of primitive hematopoietic progenitors remains elusive. FoxO’s function is restrained by AKT serine threonine protein kinase. AKT supports growth, survival and proliferation by promoting inhibition of FoxO and activation of the mammalian target of rapamycin (mTOR) and its downstream target p70 S6 Kinase (S6K) through phosphorylation. We demonstrate that loss of FoxO3 leads to a myeloproliferative-like syndrome characterized by leukocytosis, splenomegaly, enhanced generation of primitive progenitors including colony-forming-unit-spleen (CFU-S) in hematopoietic organs and hypersensitivity of hematopoietic progenitor cells to cytokines in FoxO3 null mice. These findings were intriguing since we had not found FoxO3 null hematopoietic stem cells to exhibit enhanced cycling in vivo or to generate excessive hematopoietic progenitors ex vivo (Yalcin et al., JBC, 2008). To investigate the mechanism of enhanced myeloproliferation, we interrogated cytokine-mediated activation of signaling pathways in freshly isolated FoxO3 null versus wild type bone marrow cells enriched for hematopoietic progenitors. To our surprise we found that stimulation with cytokines including IL-3 led to hyperphosphorylation of AKT, mTOR and S6K but not STAT5 proteins in FoxO3 null as compared to wild type cytokine-starved hematopoietic progenitors. In agreement with these results, in vivo administration of the mTOR inhibitor rapamycin resulted in significant reduction of FoxO3 null- but not wild type-derived CFU-Sd12 in lethally irradiated hosts. These unexpected results suggested that AKT/mTOR signaling pathway is specifically overactivated as part of a feedback loop mechanism and mediates enhanced generation of FoxO3 null primitive multipotential hematopoietic progenitors in vivo. We further showed that phosphorylation of AKT/mTOR/S6K is highly sensitive to ROS scavenger N-Acetyl-Cysteine (NAC) in vivo and ex vivo in both wild type and FoxO3 null primitive hematopoietic progenitors indicating that ROS are involved in cytokine signaling in primary hematopoietic progenitor cells. Interestingly, in vivo administration of NAC normalized the number of FoxO3 null-derived CFU-Sd12 in lethally irradiated hosts without any impact on wild type CFU-Sd12 strongly suggesting that ROS mediate specifically enhanced generation of primitive hematopoietic progenitors in FoxO3 null mice. In this context, we were surprised to find similar levels of ROS concentrations in FoxO3 mutant as compared to control hematopoietic progenitors. Thus, we asked whether the increase in FoxO3 null primitive hematopoietic progenitor compartment is due to an increase sensitivity of cytokine signaling to ROS as opposed to increased ROS build up per se in these cells. In search for a mechanism we found the expression of Lnk, a negative regulator of cytokine signaling, to be highly reduced in FoxO3 null primitive hematopoietic progenitor cells. We further demonstrated that retroviral reintroduction of Lnk but not vector control in FoxO3 null primitive bone marrow cells reduced significantly the number of FoxO3 null-derived CFU-Sd12in vivo. Collectively, these results suggest that reduced expression of Lnk hypersensitizes FoxO3-deficient hematopoietic progenitors to ROS generated by cytokine signaling leading to myeloproliferation. These cumulative findings uncover a mechanism by which deregulation of cellular sensitivity to physiological ROS leads to hematopoietic malignancies specifically in disorders in which FoxO play a role.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3847-3847
Author(s):  
Vladan P Cokic ◽  
Dragana Markovic ◽  
Olivera Mitrovic ◽  
Sanja Vignjevic ◽  
Dragoslava Djikic ◽  
...  

Abstract Abstract 3847 The microvessel density of bone marrow is increased in myeloproliferative neoplasms (MPN) parallel with vascular endothelial growth factor (VEGF). VEGF-mediated angiogenesis requires nitric oxide (NO) production from activated endothelial NO synthase (eNOS). NO as well as hypoxia stimulate the VEGF gene expression and angiogenesis by enhancing hypoxia inducible factor (HIF)-1 activity. We studied 126 newly diagnosed patients with BCR-ABL− MPN: 64 polycythemia vera (PV), 36 essential thrombocythemia (ET), 26 primary myelofibrosis (PMF) and 12 healthy individuals. We performed a combined analysis of hematopoietic CD34+ progenitor cells and granulocytes in peripheral blood of these individuals. The eNOS protein level is more than three-fold elevated in granulocytes of JAK2V617F homozygous PV patients. The essential inducer of angiogenesis VEGF-A has also about three-fold elevation at the protein level in granulocytes of PV patients, with major increases in JAK2V617F homozygous forms. Immunohistochemical analysis reveal that the percentage of VEGF-A-positive cells is increased in bone marrow of PV (5.58±0.7%) compared to normal controls (2.78±0.7%) and VEGF-A mRNA levels are increased in hematopoietic progenitor cells of PV origin. Transcription factor HIF-1α gene expression is decreased in hematopoietic progenitor cells and increased in granulocytes of PV patients. Negative regulator of HIF-1α activity, a transcription factor HIF-3α, has decreased expression in hematopoietic progenitor cells and not changed in granulocytes. In contrast to PV patients, PMF and ET disorders with a minor JAK2 mutation burden demonstrate reduced eNOS and VEGF protein levels and decreased HIF-1a gene expression in peripheral blood granulocytes, although the increase in percentage of VEGF-A-positive cells in bone marrow observed in PV patients is also evident. The present results expand the significance of JAK2V617F mutation in induction of angiogenic factors eNOS and VEGF in granulocytes of PV patients with enhanced HIF-1α presence. Moreover, the stromal and hematopoietic cells also show increased VEGF protein expression in bone marrow of PV patients. Therefore, we find that variations in angiogenic factors expression among MPN patients appear to be related to JAK2V617F mutation allele burden. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2421-2421 ◽  
Author(s):  
Cheng Li ◽  
Daniel R. George ◽  
Nichole M. Helton ◽  
Jeffery M. Klco ◽  
Jacqueline L. Mudd ◽  
...  

Abstract We have previously reported a method to produce early hematopoietic progenitor cells from C57BL/6J-derived mouse embryonic stem cells (mESCs). After co-culture on OP9 stromal cells for one week, four different C57Bl/6 mESC lines consistently differentiated into hematopoietic progenitors, as determined by immunophenotyping; we detected cells that mark as KLS (Lin- Kit+ Sca+), CMPs, GMPs, and MEPs (but not SLAMs) from all four lines. In addition, functional progenitors for erythrocytes, monocytes, and mast cells (by morphology and immunophenotyping) were detected after another week of culture in methylcellulose with hematopoietic cytokines (SCF, IL-3, IL-6, and Epo). These findings were replicated using four different lots of fetal bovine serum, and with three different lots of OP9 cells from ATCC. We injected 1x106 “OP9-induced” progenitor cells retroorbitally into unconditioned NSG mice, and detected multilineage hematopoietic engraftment (myeloid compartments marked by CD34, CD11b, Kit, and Gr-1, lymphoid compartments marked by CD3 and B220, and erythroid compartments marked by Ter119) in the bone marrow and/or spleens of 10 out of 19 recipients at 3 months. Using the OP9 co-culture system, we have differentiated miPSC clones from three independent iPSC experiments, using an integrating polycistronic lentivirus expressing OCT4, SOX2, and KLF4 as the reprogramming vector. One set of miPSC clones was produced from mouse embryonic fibroblasts (MEFs) from pooled C57BL/6J embryos, and two sets were made from adult mouse fibroblasts derived from a single animal, producing 6, 12, and 12 independent iPS clones for analysis, respectively. All thirty clones had pluripotent features, as determined by alkaline phosphatase staining and immunophenotyping (SSEA1, Oct4, and Nanog). We have injected the OP9-induced progenitors derived from one miPSC clone into NSG mice; thus far, 2 out of 14 recipients have demonstrated engraftment in the peripheral blood. However, the efficiency of hematopoietic progenitor generation with OP9 induction (based on the immunophenotyping and progenitor assays noted above) was highly variable for miPSCs from all three experiments. Among all three sets of miPSC clones, 18/30 exhibited differentiation efficiencies comparable to wild-type B6 ESCs, 5/30 clones exhibited moderately reduced differentiation efficiencies, 5/30 clones exhibited markedly reduced differentiation efficiencies, and 2/30 clones (from two different iPSC experiments) did not produce any detectable hematopoietic progenitors with OP9 induction. These phenotypes were stable and highly reproducible. The 2 clones that did not yield any hematopoietic progenitors had robust pluripotency marks, and one that was injected into the hindflank of NSG mice produced cystic teratomas. We found that 2% DMSO pretreatment of mESCs for 24 hours prior to OP9 co-culture improved the differentiation efficiency of wild-type B6 ESCs by 50% (Chetty et al. Nature Methods 10(6):553-6, 2013), but it did not rescue the phenotype of miPSC clones that did not produce hematopoietic progenitors. We are currently performing exome sequencing on the 24 miPSC clones from the adult fibroblast reprogramming experiments to determine whether phenotypic heterogeneity is due to specific mutations in the iPSC clones (Young et al. Cell Stem Cell 10(5):570-82, 2012). In summary, we have developed a simple system to derive functional early hematopoietic progenitor cells from mouse embryonic stem cells and/or induced pluripotent stem cells. OP9-induced progenitor cells engraft into NSG mice without the need for forced expression of HoxB4 (Wang et al. Proc Natl Acad Sci USA 102(52):19081-6, 2005). We have detected functional heterogeneity in miPSC clones derived from the same parental cells, which could be due to genetic variation in the founding cell from which each clone was derived, to different integration sites of the OSK lentivirus in each clone, or to as yet undefined epigenetic mechanisms. Exome sequencing may help to resolve this issue. Regardless, this approach could be a valuable tool for studying the hematopoietic development of a variety of mESC lines and/or miPSC lines derived from genetically altered mice. Disclosures: No relevant conflicts of interest to declare.


2002 ◽  
Vol 195 (2) ◽  
pp. 151-160 ◽  
Author(s):  
Satoshi Takaki ◽  
Hatsue Morita ◽  
Yoshinari Tezuka ◽  
Kiyoshi Takatsu

Hematopoietic stem cells (HSCs) give rise to variety of hematopoietic cells via pluripotential progenitors and lineage-committed progenitors and are responsible for blood production throughout adult life. Amplification of HSCs or progenitors represents a potentially powerful approach to the treatment of various blood disorders and to applying gene therapy by bone marrow transplantation. Lnk is an adaptor protein regulating the production of B cells. Here we show that Lnk is also expressed in hematopoietic progenitors in bone marrow, and that in the absence of Lnk, the number and the hematopoietic ability of progenitors are significantly increased. Augmented growth signals through c-Kit partly contributed to the enhanced hematopoiesis by lnk−/− cells. Lnk was phosphorylated by and associated with c-Kit, and selectively inhibited c-Kit–mediated proliferation by attenuating phosphorylation of Gab2 and activation of mitogen-activated protein kinase cascade. These observations indicate that Lnk plays critical roles in the expansion and function of early hematopoietic progenitors, and provide useful clues for the amplification of hematopoietic progenitor cells.


Blood ◽  
2013 ◽  
Vol 122 (13) ◽  
pp. 2195-2204 ◽  
Author(s):  
Christopher C. Nixon ◽  
Dimitrios N. Vatakis ◽  
Scott N. Reichelderfer ◽  
Dhaval Dixit ◽  
Sohn G. Kim ◽  
...  

Key PointsSome CD34+CD38+ intermediate hematopoietic progenitor cells express HIV-1 entry receptors and are susceptible to direct infection by HIV. Blood progenitors from HIV-exposed, humanized BLT mice show impaired hematopoietic potential and give rise to progeny that harbor provirus.


Development ◽  
2002 ◽  
Vol 129 (8) ◽  
pp. 2003-2013 ◽  
Author(s):  
Maria Teresa Mitjavila-Garcia ◽  
Michel Cailleret ◽  
Isabelle Godin ◽  
Maria Manuela Nogueira ◽  
Karine Cohen-Solal ◽  
...  

In this study, we have characterized the early steps of hematopoiesis during embryonic stem cell differentiation. The immunophenotype of hematopoietic progenitor cells derived from murine embryonic stem cells was determined using a panel of monoclonal antibodies specific for hematopoietic differentiation antigens. Surprisingly, the CD41 antigen (αIIb integrin, platelet GPIIb), essentially considered to be restricted to megakaryocytes, was found on a large proportion of cells within embryoid bodies although very few megakaryocytes were detected. In clonogenic assays, more than 80% of all progenitors (megakaryocytic, granulo-macrophagic, erythroid and pluripotent) derived from embryoid bodies expressed the CD41 antigen. CD41 was the most reliable marker of early steps of hematopoiesis. However, CD41 remained a differentiation marker because some CD41– cells from embryoid bodies converted to CD41+ hematopoietic progenitors, whereas the inverse switch was not observed. Immunoprecipitation and western blot analysis confirmed that CD41 was present in cells from embryoid bodies associated with CD61 (β3 integrin, platelet GPIIIa) in a complex. Analysis of CD41 expression during ontogeny revealed that most yolk sac and aorta-gonad-mesonephros hematopoietic progenitor cells were also CD41+, whereas only a minority of bone marrow and fetal liver hematopoietic progenitors expressed this antigen. Differences in CD34 expression were also observed: hematopoietic progenitor cells from embryoid bodies, yolk sac and aorta-gonad-mesonephros displayed variable levels of CD34, whereas more than 90% of fetal liver and bone marrow progenitor cells were CD34+. Thus, these results demonstrate that expression of CD41 is associated with early stages of hematopoiesis and is highly regulated during hematopoietic development. Further studies concerning the adhesive properties of hematopoietic cells are required to assess the biological significance of these developmental changes.


Sign in / Sign up

Export Citation Format

Share Document