scholarly journals Lymphoid-biased hematopoietic stem cells and myeloid-biased hematopoietic progenitor cells have radioprotection activity

Blood Science ◽  
2021 ◽  
Vol Publish Ahead of Print ◽  
Author(s):  
Shanshan Zhang ◽  
Aled O’Neill ◽  
Miner Xie ◽  
Peng Wu ◽  
Xiaofang Wang ◽  
...  
Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5052-5052
Author(s):  
Lina Wang ◽  
Wenli Feng ◽  
Xiao Yang ◽  
Feifei Yang ◽  
Rong Wang ◽  
...  

Abstract Ubiquitination is a posttranslational mechanism that controls diverse cellular processes. Fbxw11, a constituent of the SCF (Skp1-Cul1-F-box) ubiquitin ligase complex, targets for degradation of several important transcription factors, including NF-κB, β-cantenin and ATF4. Fbxw11 may play pivotal roles in many aspects of hematopoiesis through regulating various signal transduction pathways. However, the role of Fbxw11 on HSCs quiescence remains largely unknown. In this study, we cloned three transcript variants (Fbxw11a, Fbxw11c and Fbxw11d) to study the biological function of Fbxw11 in hematopoiesis. To elucidate Fbxw11 distribution in hematopoietic system, hematopoietic cell populations at different stages of differentiation were isolated from bone marrow of 8 week-old mice and Fbxw11 expression was studied by real-time PCR. Expression of Fbxw11 were lower in purified long term hematopoietic stem cells (LT-HSC, LSK CD34- Flt3-), but higher in short term hematopoietic stem cells (ST-HSC, LSK CD34+ Flt3-), hematopoietic stem and progenitor cells (LSK), and various hematopoietic progenitor cells. The results reveal that Fbxw11 is preferentially expressed in more mature progenitor cells. The expression of Fbxw11 in mature blood cells was also studied showing that Fbxw11 was expressed at lower level in neutrophils, higher level in B and T lymphocytes, and moderate level in monocytes. To assess the impact of Fbxw11 on reconstitution capacity of LT-HSCs, we cloned Fbxw11a, Fbxw11c and Fbxw11d into retrovirus system, respectively. LSK cells were infected with MSCV-Fbxw11a/Fbxw11c/Fbxw11d-IRES-GFP or the blank control vector MSCV-GFP. Competitive repopulation assays we performed 48h later after infection, and reconstitution in peripheral blood (PB) was analyzed every 4 weeks. Repopulation of donor cells expressing high level of Fbxw11 variants was significantly lower than those infected with control vector at 1 and 4 months in PB and at 4 months in BM after transplantation. These data indicate that Fbxw11 is negative for the long-term repopulating capacity of HSCs. To further confirm the effects of Fbxw11 variants in hematopoiesis, the effect of Fbxw11 variants on the growth and enumeration of hematopoietic progenitor cells was detected by colony-forming cell assay (CFC). The number of CFU-G, CFU-GM, CFU-GEMM and the total number of CFU were lower in LSK over-expressing Fbxw11 variants when compared with LSK control. To determine the cell-cycle distribution of HSC cells, Hoechst 33342 and Ki67 staining were performed showing that G0 phase LSK cells were decreased when they over-expressing Fbxw11 variants. In conclusion, our data reveal unrecognized roles for Fbxw11 in the regulation of HSPCs. Our findings suggest that Fbxw11 variants have negative effect on reconstitution capacity of LT-HSCs. Fbxw11 variants decrease the reconstitution capacity through promoting cell proliferation, which results in loss of hematopoietic stem cell quiescence. We anticipate that our experiments will facilitate the understanding of hematopoiesis through which Fbxw11-mediated signals control HSC quiescence and functions. The work was supported by the Grants 81300376, 81370634, 81570153 from the National Natural Science Foundation of China (NSFC); 14JCQNJC10600 from the Tianjin Science and Technology Programs; Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2702-2702
Author(s):  
Vittoria Guerini ◽  
Orietta Spinelli ◽  
Anna Salvi ◽  
Guido Finazzi ◽  
Tiziano Barbui ◽  
...  

Abstract Background In Polycythemia Vera (PV) and Essential Thrombocythemia (ET) hematopoietic progenitor cells can proliferate in vitro in the absence of exogenous growth factors. A somatic point mutation in the JAK2 gene (JAK2V617F) has been recently recognised as the key pathogenetic lesion of these diseases leading to constitutive tyrosine phosphorylation of JAK2, cytokine hypersensitivity and autonomous outgrowth of hematopoietic progenitor cells. Hystone-Deacetylase inhibitors (HDACi) are known inducers of cell differentiation, apoptosis and cell cycle arrest of neoplastic cells. ITF2357 is a new HDACi (Italfarmaco, Milano, Italy) which, at low micromolar concentration in vitro, inhibits the secretion of several cytokines such as IL-1, IL-6, VEGF and IFN-g and exerts a potent anti tumor activity against multiple myeloma (MM) and acute myeloid leukemia cells (AML) (Golay et al., submitted). ITF2357 is well tolerated when given to normal healthy volounteers and Phase II clinical trials are currently ongoing in AML and MM. Aim To investigate the ability of ITF2357 and the prototypic HDAC inhibitor Suberoyl Anilide Hydroxamic Acid (SAHA) used as control, to inhibit the spontaneous outgrowth of hematopoietic stem cells obtained from patients with PV (n= 6, all JAK2V617F ), ET (n= 13, 7 JAK2V617F ) and Idiopathic Erythrocytosis (IE, n= 6, all negative for JAK2V617F ). Results Endogenous erythroid colonies (EEC) assays were performed using mononuclear cells (MNC) from peripheral blood samples obtained from patients at the time of regular follow-up visits in our clinic. MNC obtained from IE or ET patients negative for JAK2V617F neither exhibited spontaneous EEC formation nor Epo hypersensitivity (from 0.1 UI/ml up to 10UI/ml). On the contrary, MNC from JAK2V617F PV and ET patients invariably sustained the spontaneous EEC outgrowth with a marked Epo hypersensitivity. When ITF2357 was added to the colony assay (ranging from 0.001 to 0.75 μM), a 90% inhibition of EEC formation was observed in all JAK2V617F PV and ET patients at 0.01 μM concentration, which corresponds to a blood level easily attained following oral administration of safe doses of ITF2357 to healthy individuals. By contrast, the prototypic HDAC inhibitor SAHA displayed a similar inhibitory activity on EEC formation only when used at 0.25 μM. By flow cytometry experiments performed on mature granulocytes isolated from PV patients we could show that ITF2357 does not modulate the overexpression of Leucocyte Alkaline Phospatase and CD177 (the PRV-1 gene product) thus suggesting that the inhibitory activity on hematopoietic cells is mainly due to a direct action on the stem cell compartment. Conclusion ITF2357, at concentration easily attained after low oral doses of the drug, show a potent inhibitory activity on the autonomous proliferation of hematopoietic stem cells of PV and TE carrying the JAK2 V617F mutation. This may provide the framework for a Phase II study of ITF2357 in these malignancies.


Blood ◽  
1993 ◽  
Vol 81 (3) ◽  
pp. 801-807 ◽  
Author(s):  
T Leemhuis ◽  
D Leibowitz ◽  
G Cox ◽  
R Silver ◽  
EF Srour ◽  
...  

Chronic myeloid leukemia (CML) is a malignant disorder of the hematopoietic stem cell. It has been shown that normal stem cells coexist with malignant stem cells in the bone marrow of patients with chronic-phase CML. To characterize the primitive hematopoietic progenitor cells within CML marrow, CD34+DR- and CD34+DR+ cells were isolated using centrifugal elutriation, monoclonal antibody labeling, and flow cytometric cell sorting. Polymerase chain reaction analysis of RNA samples from these CD34+ subpopulations was used to detect the presence of the BCR/ABL translocation characteristic of CML. The CD34+DR+ subpopulation contained BCR/ABL(+) cells in 11 of 12 marrow samples studied, whereas the CD34+DR- subpopulation contained BCR/ABL(+) cells in 6 of 9 CML marrow specimens. These cell populations were assayed for hematopoietic progenitor cells, and individual hematopoietic colonies were analyzed by PCR for their BCR/ABL status. Results from six patients showed that nearly half of the myeloid colonies cloned from CD34+DR- cells were BCR/ABL(+), although the CD34+DR- subpopulation contained significantly fewer BCR/ABL(+) progenitor cells than either low-density bone marrow (LDBM) or the CD34+DR+ fraction. These CD34+ cells were also used to establish stromal cell-free long-term bone marrow cultures to assess the BCR/ABL status of hematopoietic stem cells within these CML marrow populations. After 28 days in culture, three of five cultures initiated with CD34+DR- cells produced BCR/ABL(-) cells. By contrast, only one of eight cultures initiated with CD34+DR+ cells were BCR/ABL(-) after 28 days. These results indicate that the CD34+DR- subpopulation of CML marrow still contains leukemic progenitor cells, although to a lesser extent than either LDBM or CD34+DR+ cells.


Blood ◽  
1993 ◽  
Vol 81 (3) ◽  
pp. 801-807 ◽  
Author(s):  
T Leemhuis ◽  
D Leibowitz ◽  
G Cox ◽  
R Silver ◽  
EF Srour ◽  
...  

Abstract Chronic myeloid leukemia (CML) is a malignant disorder of the hematopoietic stem cell. It has been shown that normal stem cells coexist with malignant stem cells in the bone marrow of patients with chronic-phase CML. To characterize the primitive hematopoietic progenitor cells within CML marrow, CD34+DR- and CD34+DR+ cells were isolated using centrifugal elutriation, monoclonal antibody labeling, and flow cytometric cell sorting. Polymerase chain reaction analysis of RNA samples from these CD34+ subpopulations was used to detect the presence of the BCR/ABL translocation characteristic of CML. The CD34+DR+ subpopulation contained BCR/ABL(+) cells in 11 of 12 marrow samples studied, whereas the CD34+DR- subpopulation contained BCR/ABL(+) cells in 6 of 9 CML marrow specimens. These cell populations were assayed for hematopoietic progenitor cells, and individual hematopoietic colonies were analyzed by PCR for their BCR/ABL status. Results from six patients showed that nearly half of the myeloid colonies cloned from CD34+DR- cells were BCR/ABL(+), although the CD34+DR- subpopulation contained significantly fewer BCR/ABL(+) progenitor cells than either low-density bone marrow (LDBM) or the CD34+DR+ fraction. These CD34+ cells were also used to establish stromal cell-free long-term bone marrow cultures to assess the BCR/ABL status of hematopoietic stem cells within these CML marrow populations. After 28 days in culture, three of five cultures initiated with CD34+DR- cells produced BCR/ABL(-) cells. By contrast, only one of eight cultures initiated with CD34+DR+ cells were BCR/ABL(-) after 28 days. These results indicate that the CD34+DR- subpopulation of CML marrow still contains leukemic progenitor cells, although to a lesser extent than either LDBM or CD34+DR+ cells.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1234-1234
Author(s):  
Robert S Welner ◽  
Giovanni Amabile ◽  
Deepak Bararia ◽  
Philipp B. Staber ◽  
Akos G. Czibere ◽  
...  

Abstract Abstract 1234 Specialized bone marrow (BM) microenvironment niches are essential for hematopoietic stem and progenitor cell maintenance, and recent publications have focused on the leukemic stem cells interaction and placement within those sites. Surprisingly, little is known about how the integrity of this leukemic niche changes the normal stem and progenitor cells behavior and functionality. To address this issue, we started by studying the kinetics and differentiation of normal hematopoietic stem and progenitor cells in mice with Chronic Myeloid Leukemia (CML). CML accounts for ∼15% of all adult leukemias and is characterized by the BCR-ABL t(9;22) translocation. Therefore, we used a novel SCL-tTA BCR/ABL inducible mouse model of CML-chronic phase to investigate these issues. To this end, BM from leukemic and normal mice were mixed and co-transplanted into hosts. Although normal hematopoiesis was increasingly suppressed during the disease progression, the leukemic microenvironment imposed distinct effects on hematopoietic progenitor cells predisposing them toward the myeloid lineage. Indeed, normal hematopoietic progenitor cells from this leukemic environment demonstrated accelerated proliferation with a lack of lymphoid potential, similar to that of the companion leukemic population. Meanwhile, the leukemic-exposed normal hematopoietic stem cells were kept in a more quiescent state, but remained functional on transplantation with only modest changes in both engraftment and homing. Further analysis of the microenvironment identified several cytokines that were found to be dysregulated in the leukemia and potentially responsible for these bystander responses. We investigated a few of these cytokines and found IL-6 to play a crucial role in the perturbation of normal stem and progenitor cells observed in the leukemic environment. Interestingly, mice treated with anti-IL-6 monoclonal antibody reduced both the myeloid bias and proliferation defects of normal stem and progenitor cells. Results obtained with this mouse model were similarly validated using specimens obtained from CML patients. Co-culture of primary CML patient samples and GFP labeled human CD34+CD38- adult stem cells resulted in selective proliferation of the normal primitive progenitors compared to mixed cultures containing unlabeled normal bone marrow. Proliferation was blocked by adding anti-IL-6 neutralizing antibody to these co-cultures. Therefore, our current study provides definitive support and an underlying crucial mechanism for the hematopoietic perturbation of normal stem and progenitor cells during leukemogenesis. We believe our study to have important implications for cancer prevention and novel therapeutic approach for leukemia patients. We conclude that changes in cytokine levels and in particular those of IL-6 in the CML microenvironment are responsible for altered differentiation and functionality of normal stem cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2002 ◽  
Vol 100 (1) ◽  
pp. 327-333 ◽  
Author(s):  
Gerjo A. Velders ◽  
Johannes F. M. Pruijt ◽  
Perry Verzaal ◽  
Ronald van Os ◽  
Yvette van Kooyk ◽  
...  

Abstract The β2 integrins leukocyte function antigen-1 (LFA-1, CD11a) and macrophage antigen-1 (Mac-1, CD11b) have been reported to play a role in the attachment of CD34+ cells to stromal cells in the bone marrow. When administered prior to interleukin-8 (IL-8), anti–LFA-1 antibodies completely prevent the IL-8–induced mobilization of hematopoietic stem cells in mice. Here, we studied the role of anti-β2 integrin antibodies in granulocyte colony-stimulating factor (G-CSF)–induced mobilization of hematopoietic progenitor cells. Administration of antibodies against the α chain of LFA-1 or against the α chain of Mac-1 followed by daily injections of G-CSF for more than 1 day resulted in a significant enhancement of mobilization of hematopoietic progenitor cells when compared with mobilization induced by G-CSF alone. Also, the number of late (day 28) cobblestone area–forming cells in vitro was significantly higher after mobilization with anti–LFA-1 antibodies followed by 5 μg G-CSF for 5 days than with G-CSF alone (119 ± 34 days vs 17 ± 14 days), indicating mobilization of repopulating stem cells. Pretreatment with blocking antibodies to intercellular adhesion molecule-1 (ICAM-1; CD54), a ligand of LFA-1 and Mac-1, did not result in an effect on G-CSF–induced mobilization, suggesting that the enhancing effect required an interaction of the β2 integrins and one of their other ligands. Enhancement of mobilization was not observed in LFA-1–deficient (CD11a) mice, indicating that activated cells expressing LFA-1 mediate the synergistic effect, rather than LFA-1–mediated adhesion.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1499-1499
Author(s):  
Pratibha Singh ◽  
Jonathan Hoggatt ◽  
Jennifer M. Speth ◽  
Louis M. Pelus

Abstract Abstract 1499 Poster Board I-522 Dendritic cells (DCs) are an attractive target for therapeutic manipulation of the immune system due to their potent antigen presentation capacity and ability to induce effective immune response. In steady-state conditions different DC subsets including myeloid DCs (CD11c+CD11b+B220neg) and plasmacytoid DCs (CD11c+CD11bnegB220+) are generated in bone-marrow (BM) from hematopoietic stem cells through a series of differentiation steps. We recently demonstrated that prostaglandin (PGE2), the predominant metabolite of arachidonic acid metabolism by cyclooxygenase (COX) enzymes, enhances the homing, survival and proliferation of hematopoietic stem cells (Hoggatt et. al., Blood 2009). In this study, we examined the requirement of prostaglandins in development of DCs from hematopoietic progenitor cells. In vivo treatment of mice for 4 days with the non-steroidal anti-inflammatory drug (NSAID) indomethacin (2.5 mg/kg/bid), a dual COX1/COX2 inhibitor, produced a 59.5±17.8 % (p<0.02) reduction in total bone-marrow DC number compared to control mice treated with vehicle alone. Interestingly, indomethacin selectively decreased marrow CD11c+CD11b+B220neg myeloid DCs without affecting CD11c+CD11bnegB220+ plasmacytoid DCs. To determine whether fewer DCs in the bone marrow of indomethacin treated mice was due to the impairment of DC differentiation from their hematopoietic progenitor cells, we stimulated differentiation of DCs from lineage depleted (CD5−,CD45R−, CD11b−, Anti-Gr-1−, 7-4− and Ter-119−) bone marrow cells using Flt-3 ligand for 9 days in vitro and treated cultures with indomethacin (1 microM), SC-560 (10 microM), a selective COX-1 inhibitor or NS-398 (10 microM), a selective COX-2 inhibitor. Indomethacin produced a 1.98±0.38 fold, (p<0.02) reduction and the COX-2 inhibitor NS-398 produced a 1.52±0.04 fold, (p<0.05) reduction in CD11c+CD11b+B220neg myeloid DC generation compared to control, while the COX-1 specific inhibitor SC-560 was without effect. As expected, Flt-3-ligand induced plasmacytoid DC (CD11c+CD11bnegB220+) differentiation was not affected by selective COX inhibitors. Indomethacin also impaired generation of CD11a+CD14neg Langerhans DC from human umbilical cord blood CD34+ cells. Measurement of PGE2 production in culture supernatants from DC-producing cultures demonstrated detectable PGE2 after 6 days of culture and DC generation from BM progenitors in these cultures was impaired when PGE2 synthesis was blocked on day 6 by indomethacin administration. Indomethacin treatment during the first 5 days of Flt3-ligand stimulated DC differentiation cultures did not decrease DC production. To identify mechanisms responsible for this impairment in Flt-3 ligand-induced DC generation from hematopoietic progenitor cells, we analyzed the effect of indomethacin on DC-committed precursor cell proliferation and survival. Survival of DC-committed precursors defined as CD11clow CD11bbrightMHCIIlow was reduced 35±2.5% (p<0.05) in indomethacin treated cultures compared to control. However, indomethacin did not affect DC precursor proliferation as measured by BrdU incorporation assay. To elucidate the signaling mechanisms by which indomethacin impaired the survival of DC precursors, we added selective receptor agonists to each of known PGE receptors, EP1-4, during Flt3-ligand induced DC differentiation. The EP1/EP3 agonist 17-phenyl trinor PGE2 rescued the DC precursors from indomethacin mediated death, whereas Butaprost, a specific EP2 agonist and L902688, a selective EP4 agonist, failed to rescue DC precursor death. DCs developed in the presence of prostaglandin inhibitors did not show any defect in LPS-induced activation and expressed CD40, CD80, CD86 and MHCII levels similar to control as measured by flow cytometry. In addition, DC developed in the absence of endogenous PGE2 production successfully induced T-cell activation as measured by mix lymphocyte reaction assay (MLR). In conclusion, COX-2 mediated prostaglandin production by DC-committed hematopoietic precursors confers resistance to cell death via signaling through EP1/EP3 receptors and promotes dendritic cell development. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (2) ◽  
pp. 290-298 ◽  
Author(s):  
Anja Köhler ◽  
Vince Schmithorst ◽  
Marie-Dominique Filippi ◽  
Marnie A. Ryan ◽  
Deidre Daria ◽  
...  

Abstract Aged hematopoietic stem cells (HSCs) are impaired in supporting hematopoiesis. The molecular and cellular mechanisms of stem cell aging are not well defined. HSCs interact with nonhematopoietic stroma cells in the bone marrow forming the niche. Interactions of hematopoietic cells with the stroma/microenvironment inside bone cavities are central to hematopoiesis as they regulate cell proliferation, self-renewal, and differentiation. We recently hypothesized that one underlying cause of altered hematopoiesis in aging might be due to altered interactions of aged stem cells with the microenvironment/niche. We developed time-lapse 2-photon microscopy and novel image analysis algorithms to quantify the dynamics of young and aged hematopoietic cells inside the marrow of long bones of mice in vivo. We report in this study that aged early hematopoietic progenitor cells (eHPCs) present with increased cell protrusion movement in vivo and localize more distantly to the endosteum compared with young eHPCs. This correlated with reduced adhesion to stroma cells as well as reduced cell polarity upon adhesion of aged eHPCs. These data support a role of altered eHPC dynamics and altered cell polarity, and thus altered niche biology in mechanisms of mammalian aging.


Sign in / Sign up

Export Citation Format

Share Document