NF-kB Family Proteins Participate in Multiple Steps of Hematopoiesis through Elimination of Reactive Oxygen Species (ROS).

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1694-1694
Author(s):  
Soichi Nakata ◽  
Itaru Matsumura ◽  
Hirokazu Tanaka ◽  
Yusuke Satoh ◽  
Takumi Era ◽  
...  

Abstract NF-kB family proteins have been reported to induce the expression of over 150 target genes, thereby crucially regulating immune responses, stress responses, and inflammation. These proteins also play important roles in cell growth and survival in various cell types. However, the precise roles of NF-kB in hematopoiesis and their mechanisms remain undetermined. To examine the roles for NF-kB family proteins in the growth and survival of hematopoietic cells, we expressed dominant negative NF-kB (IkBSR) in a murine IL-3-dependent cell line Ba/F3 using a Lac-inducible system, in which IkBSR was inducibly expressed by the IPTG treatment; this clone was designated Ba/F3/IkBSR. Furthermore, we introduced EPO receptor (R), TPOR, and G-CSFR/gp130 consisting of the extracelluar domain of G-CSFR and cytoplasmic domain of gp130 into this clone. At first, we confirmed that these clones could survive and proliferate under the cultures with IL-3, EPO, TPO, G-CSF, respectively. Although IPTG-induced IkBSR slightly suppressed IL-3- and EPO-dependent growth at low concentrations, it did not affect TPO- or gp130L-dependent growth, suggesting that NF-kB might not be so important for cytokine-dependent growth of hematopoietic cells. In contrast, IkBSR prominently enhanced factor-deprived apoptosis, which was accompanied by the ROS accumulation. To access the roles of ROS in IkBSR-enhanced apoptosis, we overexpressed ROS scavenger enzymes MnSOD and thioredoxin X (TRX) in Ba/F3/IkBSR, respectively. As a result, MnSOD and TRX significantly canceled IkB-SR-enhanced apoptosis, suggesting that ROS would be responsible for this apoptosis. We next analyzed the effects of IkBSR on the growth and survival of normal hematopoietic cells. When IkBSR was introduced into murine Lin−Sca-1+ hematopoietic stem/progenitor cells with the retrovirus system, it induced apoptosis even in the presence of appropriate cytokines. This apoptosis was also accompanied by the ROS accumulation due to the downregulated expression of anti-oxidants such as glutathione, MnSOD, glutathione peroxidase, and TRX. In addition, the expression of antiapoptotic BCl-2 family members, Bcl-XL, Bcl-2, and A1 was found to be repressed by IkBSR. However, since antioxidants such as MCI (3-methyl-1-phenyl-2-pyrazolin-5-one), N-acetylecysteine and TRX cancelled this apoptosis, ROS were supposed to be more important for IkBSR-induced apoptosis in normal hematopoietic stem/progenitor cells. To further analyze the roles for NF-kB proteins in the development of hematopoietic cells, we expressed IkBSR in an inducible fashion at various stages of hematopoiesis using the OP9 system, in which hematopoietic cells are induced to develop from ES cells. When IkBSR was expressed at the stage of hemangioblasts, IkBSR induced apoptosis and inhibited the development of hematopoietic stem cells, which was also cancelled by MCI. Furthermore, when IkBSR was expressed after the development of hematopoietic stem cells, it also inhibited terminal differentiation towards granulocytes, erythrocytes, and megakaryocytes through ROS-mediated apoptosis; IkBSR inhibited granulopoiesis before the development of myeloblasts, erythropoiesis after the development of proerythroblasts, and megakaryopoiesis during polyploidization of megakaryocytes. These results indicate that NF-kB family proteins play essential roles to prevent apoptosis at multiple steps of hematopoiesis by eliminating ROS.

2011 ◽  
Vol 9 (1) ◽  
pp. 64-73 ◽  
Author(s):  
Scott W. Boyer ◽  
Aaron V. Schroeder ◽  
Stephanie Smith-Berdan ◽  
E. Camilla Forsberg

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1320-1320
Author(s):  
Sachiko Ezoe ◽  
Itaru Matsumura ◽  
Hirokazu Tanaka ◽  
Hirohiko Shibayama ◽  
Masao Mizuki ◽  
...  

Abstract Sir2 (silent information regulator 2) initially isolated as a transcriptional silencer was shown to be essential for the longevity caused by the calorie restriction in yeast and C. elegans. Also, SIRT1, a murine homologue of Sir2, was reported to play pivotal roles in cellular senescence as a calorie sensor. SIRT1 deficiency in mice was reported to result in early postnatal lethality because of the incomplete development of embryos and/or heart malformation. However, in these studies, the abnormality in hematopoiesis was not precisely analyzed. So, in the present study, we investigated the role of SIRT1 in hematopoietic stem cells. At first, we examined the expression profile of SIRT1 in various hematopoietic cells by RT-PCR analysis, and found that it was ubiquitously expressed in all of the hematopoietic lineages from immature to differentiated cells. Next, we examined the effects of SIRT1 inhibitor, nicotinamide(NA), and its activator, resveratol, on murine hematopoietic stem/progenitor cells. We isolated Lineage(−) Sca-1(+)(LS) cells from murine bone marrow and cultured with the cytokine cocktail containing SCF, IL-6, Flt3L, and TPO, which is utilized for the expansion of stem cells, together with NA or resveratol. As a result, NA significantly reduced LS cell population from 21.5% to 5.7%, while resveratol increased this fraction up to 37.4%. Also, we performed colony assays using LS cells cultured with or without NA for two days. The numbers of CFU-mix, BFU-E, CFU-E, CFU-G, CFU-M, and CFU-Meg yielded from NA-treated cells were all reduced about 50–80% as compared with those from untreated cells. We also examined the effects of NA on terminal differentiations of LS cells. For this purpose, we cultured LS cells using the following cytokine combinations: SCF and G-CSF for inducing granulocytic differentiation; SCF and EPO for erythroid differentiation, or with SCF and TPO for megakaryocytic differentiation. NA accelerated differentiation toward all lineages. To inhibit SIRT1 activity more specifically, we introduced SiRNA for SIRT1 into murine LS cells using the retrovirus system. In consistent with the results obtained from the experiments using NA, the proportion of immature LS cells was reduced from 8.0% to 1.8%, and terminal differentiation was promoted in SIRT1 SiRNA-infected cells in comparison with MOCK-infected cells. We further examined the mechanisms through which SIRT1 keeps hematopoietic cells undifferentiated. Since p38MAPK cascade is also reported to promote differentiation of hematopoietic stem cells, we analyzed whether p38MAPK is a downstream effector of SIRT1 using its inhibitor SB202190. Even if SB202190 was added to the culture medium of LS cells, it did not affect NA-induced acceleration of differentiation, suggesting that p38MAPK does not act as a downstream effector of SIRT1. In a recent paper, SIRT1 was reported to regulate cellular resistance against stress and apoptosis by suppressing p53 activity through its deacetylation. So, we examined the effects of NA using LS cells isolated from p53 knockout mice. However, NA accelerated differentiation of p53−/− LS cells as was the case with those from normal mice, indicating that SIRT1 suppresses the differentiation of hematopoietic stem/progenitor cells independently of p53. Thus, SIRT1 was supposed to regulate the fate and differentiation of hematopoietic cells through a novel system as a calorie sensor.


2018 ◽  
Author(s):  
Petter Säwén ◽  
Mohamed Eldeeb ◽  
Eva Erlandsson ◽  
Trine A Kristiansen ◽  
Cecilia Laterza ◽  
...  

ABSTRACTA hallmark of adult hematopoiesis is the continuous replacement of blood cells with limited lifespans. It is well established that adult hematopoietic stem cells (HSCs) are active contributors to these processes after transplantation, yet their role in native hematopoiesis has recently been called into question. Here, we use inducible lineage tracing from genetically marked adult HSCs to explore their roles in the steady state. We show that adult HSCs contribute robustly to all lineages via intermediate progenitor cells, but with neglible production of hematopoietic cells with a known fetal origin. We further reveal that the timing for regeneration of distinct blood lineages varies substantially. Finally, HSC contribution to multilineage hematopoiesis in aged animals declines with increasing age. Therefore, while HSCs are active contributors to native adult hematopoiesis, it appears that the numerical increase of HSCs is a physiologically relevant compensatory mechanism to account for a reduced differentiation capacity with age.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 10-10
Author(s):  
Rasoul Pourebrahim ◽  
Rafael Heinz Montoya ◽  
Zoe Alaniz ◽  
Lauren B Ostermann ◽  
Edward Ayoub ◽  
...  

The murine double minute 2 (Mdm2) protein is an important negative regulator of the p53 tumor suppressor, required for normal embryonic development and homeostasis. In humans, a single nucleotide polymorphism in the MDM2 promoter is associated with increased risk of cancer suggesting the importance of MDM2 levels in tumorigenesis (Bond et al., 2004). Mice with Mdm2 haploinsufficiency were previously reported as phenotypically normal with increased p53-dependent response to ionizing radiation (IR) resulting in lethal bone marrow failure (Terzian et al., 2007). However, the mechanism of radiosensitivity in these mice is unknown. To better characterize the phenotype of Mdm2 haploinsufficient mice and explore the mechanism of IR sensitivity, we developed a lineage tracing system to genetically label and trace the fate of cells after heterozygous deletion of Mdm2 in hematopoietic as well as mesenchymal progenitor cells. We utilized mTmG allele as a traceable reporter in which green fluorescence (GFP) replaces red fluorescence (TdTomato) after Cre-mediated recombination. Using Vav-Cre or Mx1-Cre, we first targeted Mdm2 in hematopoietic cells and marked them by TdTomato (Mdm2-WT) and GFP (Mdm2+/-). Heterozygous deletion of Mdm2 in hematopoietic stem cells using Vav-Cre resulted in massive apoptosis of emerging hematopoietic progenitor cells in the aorta-gonad-mesonephros (AGM) region at E10.5. Marker segregation analysis by fluorescence microscopy and flow cytometry revealed a population of hematopoietic stem cells having both TdTomato and GFP markers that escaped from apoptosis and reconstituted the hematopoietic cells in the fetal liver. Deletion of p53 in these mice did not rescue the apoptotic phenotype of hematopoietic cells with Mdm2 haploinsufficiency suggesting that a non-p53 dependent function of Mdm2 is necessary for proper development of hematopoietic stem cells in early development. In adult mice, Mdm2 haploinsufficiency in hematopoietic cells resulted in significant reduction in bone marrow hematopoietic stem cells in the absence of IR induced cellular stress. In Mx1-Cre;mTmG;Mdm2fl/+ mice, induction of Cre activity by pIpC injection resulted in hematopoietic failure evident by pancytopenia in peripheral blood. To test whether the same apoptotic response to Mdm2 haploinsufficiency can occur in other lineages, we generated a traceable conditional model of Mdm2 haploinsufficiency in mesenchymal progenitor cells using Osx-Cre and Prx1-Cre. Mice with heterozygous deletion of Mdm2 (Osx-Cre;mTmG;Mdm2fl/+) showed apoptosis of emerging osteoprogenitor cells at E16.5. Analysis of bone at 4 weeks revealed significant apoptosis of emerging osteoprogenitor cells further supporting our findings in the hematopoietic lineage. Together, our data highlights the importance of Mdm2 levels in hematopoietic and mesenchymal stem cell hemostasis and identifies depletion of hematopoietic stem cells in the bone marrow as the mechanism of radiosensitivity in Mdm2 haploinsufficient mice. Disclosures Andreeff: Daiichi-Sankyo; Breast Cancer Research Foundation; CPRIT; NIH/NCI; Amgen; AstraZeneca: Research Funding; Daiichi-Sankyo; Jazz Pharmaceuticals; Celgene; Amgen; AstraZeneca; 6 Dimensions Capital: Consultancy; Centre for Drug Research & Development; Cancer UK; NCI-CTEP; German Research Council; Leukemia Lymphoma Foundation (LLS); NCI-RDCRN (Rare Disease Clin Network); CLL Founcdation; BioLineRx; SentiBio; Aptose Biosciences, Inc: Membership on an entity's Board of Directors or advisory committees; Amgen: Research Funding.


Blood ◽  
1996 ◽  
Vol 87 (9) ◽  
pp. 3676-3687 ◽  
Author(s):  
AP Warren ◽  
K Patel ◽  
DJ McConkey ◽  
R Palacios

In our search for cell surface markers expressed on hematopoietic stem cells and/or very early progenitor cells we found that the Joro 177 monoclonal antibody (MoAb) bound to most hematopoietic cells in day 8/8.5 yolk sac, day 12 fetal liver, and day 13 fetal thymocytes; it stained hematopoietic stem cells and less immature lymphoid, myeloid, and erythroid-lineage cells, but not most thymocytes and splenic lymphocytes in adult mice. Joro 177 MoAb stimulated tyrosine phosphorylation of an integral of 124-kD protein and induced homotypic aggregation of lymphoid progenitor cells. Importantly, Joro 177 MoAb inhibited cell survival/growth and consequently the generation of lymphoid, myeloid, and erythroid lineage cells in vitro from early Lin- hematopoietic precursors. Joro 177 MoAb induced apoptosis of hematopoietic progenitor cells. Molecular cloning and expression indicated that Joro 177 MoAb recognizes a type II transmembrane protein, which is the mouse homologue of the human CD98 heavy chain gene. We suggest that CD98 is a cell membrane receptor involved in the control of cell survival/death of hematopoietic cells.


Blood ◽  
1996 ◽  
Vol 88 (10) ◽  
pp. 3774-3784 ◽  
Author(s):  
F Morel ◽  
SJ Szilvassy ◽  
M Travis ◽  
B Chen ◽  
A Galy

The CD34 antigen is expressed on most, if not all, human hematopoietic stem cells (HSCs) and hematopoietic progenitor cells, and its use for the enrichment of HSCs with repopulating potential is well established. However, despite homology between human and murine CD34, its expression on subsets of primitive murine hematopoietic cells has not been examined in full detail. To address this issue, we used a novel monoclonal antibody against murine CD34 (RAM34) to fractionate bone marrow (BM) cells that were then assayed in vitro and in vivo with respect to differing functional properties. A total of 4% to 17% of murine BM cells expressed CD34 at intermediate to high levels, representing a marked improvement over the resolution obtained with previously described polyclonal anti-CD34 antibodies. Sixty percent of CD34+ BM cells lacked lineage (Lin) markers expressed on mature lymphoid or myeloid cells. Eighty-five percent of Sca-1+Thy-1(10)Lin- /10 cells that are highly enriched in HSCs expressed intermediate, but not high, levels of CD34 antigen. The remainder of these phenotypically defined stem cells were CD34-. In vitro colony-forming cells, day-8 and -12 spleen colony-forming units (CFU-S), primitive progenitors able to differentiate into B lymphocytes in vitro or into T lymphocytes in SCID mice, and stem cells with radioprotective and competitive long-term repopulating activity were all markedly enriched in the CD34+ fraction after single-parameter cell sorting. In contrast, CD34-BM cells were depleted of such activities at the cell doses tested and were capable of only short-term B-cell production in vitro. The results indicate that a significant proportion of murine HSCs and multilineage progenitor cells express detectable levels of CD34, and that the RAM34 monoclonal antibody is a useful tool to subset primitive murine hematopoietic cells. These findings should facilitate more direct comparisons of the biology of CD34+ murine and human stem and progenitor cells.


2004 ◽  
Vol 24 (15) ◽  
pp. 6751-6762 ◽  
Author(s):  
Keita Kirito ◽  
Norma Fox ◽  
Kenneth Kaushansky

ABSTRACT Members of the homeobox family of transcription factors are major regulators of hematopoiesis. Overexpression of either HOXB4 or HOXA9 in primitive marrow cells enhances the expansion of hematopoietic stem cells (HSCs). However, little is known of how expression or function of these proteins is regulated during hematopoiesis under physiological conditions. In our previous studies we demonstrated that thrombopoietin (TPO) enhances levels of HOXB4 mRNA in primitive hematopoietic cells (K. Kirito, N. Fox, and K. Kaushansky, Blood 102:3172-3178, 2003). To extend our studies, we investigated the effects of TPO on HOXA9 in this same cell population. Although overall levels of the transcription factor were not affected, we found that TPO induced the nuclear import of HOXA9 both in UT-7/TPO cells and in primitive Sca-1+/c-kit+/Gr-1− hematopoietic cells in a mitogen-activated protein kinase-dependent fashion. TPO also controlled MEIS1 expression at mRNA levels, at least in part due to phosphatidylinositol 3-kinase activation. Collectively, TPO modulates the function of HOXA9 by leading to its nuclear translocation, likely mediated by effects on its partner protein MEIS1, and potentially due to two newly identified nuclear localization signals. Our data suggest that TPO controls HSC development through the regulation of multiple members of the Hox family of transcription factors through multiple mechanisms.


2005 ◽  
Vol 67 (1) ◽  
Author(s):  
Hideo Ema ◽  
Yohei Morita ◽  
Hiromitsu Nakauchi ◽  
Yumi Matsuzaki

Cells ◽  
2020 ◽  
Vol 9 (5) ◽  
pp. 1293 ◽  
Author(s):  
Ghmkin Hassan ◽  
Masaharu Seno

The concepts of hematopoiesis and the generation of blood and immune cells from hematopoietic stem cells are some steady concepts in the field of hematology. However, the knowledge of hematopoietic cells arising from solid tumor cancer stem cells is novel. In the solid tumor microenvironment, hematopoietic cells play pivotal roles in tumor growth and progression. Recent studies have reported that solid tumor cancer cells or cancer stem cells could differentiate into hematopoietic cells. Here, we discuss efforts and research that focused on the presence of hematopoietic cells in tumor microenvironments. We also discuss hematopoiesis from solid tumor cancer stem cells and clarify the notion of differentiation of solid tumor cancer stem cells into non-cancer hematopoietic stem cells.


Sign in / Sign up

Export Citation Format

Share Document