Direct Interaction with Macrophages Increases Proerythroblast Proliferation While Preserving Erythropoietin (EPO) Dependence.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1287-1287
Author(s):  
Melissa M. Rhodes ◽  
Prapaporn Kopsombut ◽  
Maurice Bondurant ◽  
James O. Price ◽  
Mark J. Koury

Abstract INTRODUCTION: EPO regulates erythropoiesis by preventing apoptosis at the relatively late developmental stages of CFU-E and proerythroblasts. Cells in these EPO-dependent stages are actively dividing, but after several divisions they enter a G0 state from which they enucleate. In vivo these erythroid progenitor cells associate physically with macrophages in the bone marrow, forming erythroblastic islands. Erythroblastic islands appear to be necessary for proper development of erythroblasts into erythrocytes, but our current knowledge about erythroid progenitor-macrophage interactions in the erythroblastic islands is limited. METHODS: Spleens of mice in the acute erythroblastic phase of Friend virus disease were used to reconstitute erythroblastic islands in a co-culture system that enabled study of interactions between macrophages and developmentally synchronized EPO-dependent erythroid progenitors. Proliferation and differentiation of these erythroid progenitors in macrophage co-cultures was compared to controls in which the same erythroid progenitors were cultured alone. Erythroblasts adherent to macrophages and non-adherent erythroblasts from co-cultures, as well as control erythroblasts cultured without macrophages were collected at 6, 20, 32, and 44 hrs after initial culture for cell counts, cytospin preparations for morphology, flow cytometry analyses for apoptosis (TUNEL), cell cycle phases, and expression of two surface molecules known to be expressed on differentiating erythroblasts, phosphatidylserine (PS) and α4 integrin. Experiments were also done with erythroblasts cultured in macrophage-conditioned media. RESULTS: Splenic erythroblasts cultured alone proliferated 4.6 ± 0.7 fold over 44 h, while erythroblasts co-cultured with splenic macrophages proliferated 14.2 ± 2 fold (n=12). Control erythroblasts had the same proliferation in macrophage-conditioned medium as they did in normal medium. In EPO dose-response experiments, percentages of apoptosis were the same among adherent and non-adherent co-cultured erythroblasts and control erythroblasts. Cytospin preparations revealed no differences in morphology among non-adherent and adherent erythroblasts in co-cultures and control erythroblasts. No differences were found in enucleation percentages, extruded nuclei, or reticulocyte formation at 44 h. Likewise no differences were found in percentages of apoptotic cells, distribution of cell cycle phases, or surface expressions of PS or α4 integrin during the 44 h of differentiation. CONCLUSIONS: Co-culture with macrophages in reconstituted erythroblastic islands dramatically increases the erythroblast proliferation, without affecting differentiation. The increase in proliferation is not due to decreased apoptosis, increased EPO responsiveness, or soluble factors released by the macrophages. Preservation of EPO-dependence during this expansion of erythroblasts mediated by direct interaction with macrophages indicates that erythropoietic regulation by EPO affects a larger population of erythroid progenitor cells in later stages of erythropoiesis and, thereby, accounts for relatively rapid increases or decreases in erythrocyte production following changes in EPO levels in vivo.

Blood ◽  
1991 ◽  
Vol 78 (10) ◽  
pp. 2493-2497 ◽  
Author(s):  
CH Dai ◽  
SB Krantz ◽  
KM Zsebo

Abstract To understand the factors that regulate the early growth and development of immature erythroid progenitor cells, the burst-forming units-erythroid (BFU-E), it is necessary to have both highly purified target cells and a medium free of serum. When highly purified human blood BFU-E were cultured in a serum-free medium adequate for the growth of later erythroid progenitors, BFU-E would not grow even with the addition of recombinant human interleukin-3 (rIL-3), known to be essential for these cells. However, the addition of recombinant human stem cell factor (rSCF), which supports germ cell and pluripotential stem cell growth, stimulated BFU-E to grow equally well in serum-free as in serum-containing medium. Limiting dilution studies showed that rSCF acts directly on the BFU-E that do not require accessory cells for growth. Furthermore, rSCF was necessary for BFU-E development during the initial 7 days of culture, until these cells reached the stage of the late progenitors, the colony-forming units-erythroid (CFU-E). These studies indicate that early erythropoiesis is dependent on the direct action of SCF that not only affects early stem cells but is continually necessary for the further development of committed erythroid progenitor cells until the CFU-E stage of maturation.


2008 ◽  
Vol 28 (24) ◽  
pp. 7394-7401 ◽  
Author(s):  
Andre M. Pilon ◽  
Murat O. Arcasoy ◽  
Holly K. Dressman ◽  
Serena E. Vayda ◽  
Yelena D. Maksimova ◽  
...  

ABSTRACT Erythroid Krüppel-like factor (EKLF) is a Krüppel-like transcription factor identified as a transcriptional activator and chromatin modifier in erythroid cells. EKLF-deficient (Eklf −/− ) mice die at day 14.5 of gestation from severe anemia. In this study, we demonstrate that early progenitor cells fail to undergo terminal erythroid differentiation in Eklf −/− embryos. To discover potential EKLF target genes responsible for the failure of erythropoiesis, transcriptional profiling was performed with RNA from wild-type and Eklf −/− early erythroid progenitor cells. These analyses identified significant perturbation of a network of genes involved in cell cycle regulation, with the critical regulator of the cell cycle, E2f2, at a hub. E2f2 mRNA and protein levels were markedly decreased in Eklf −/− early erythroid progenitor cells, which showed a delay in the G1-to-S-phase transition. Chromatin immunoprecipitation analysis demonstrated EKLF occupancy at the proximal E2f2 promoter in vivo. Consistent with the role of EKLF as a chromatin modifier, EKLF binding sites in the E2f2 promoter were located in a region of EKLF-dependent DNase I sensitivity in early erythroid progenitor cells. We propose a model in which EKLF-dependent activation and modification of the E2f2 locus is required for cell cycle progression preceding terminal erythroid differentiation.


Blood ◽  
1996 ◽  
Vol 88 (3) ◽  
pp. 803-808 ◽  
Author(s):  
K Carver-Moore ◽  
HE Broxmeyer ◽  
SM Luoh ◽  
S Cooper ◽  
J Peng ◽  
...  

Abstract Thrombopoietin (TPO), the ligand for the c-mpl receptor, has been shown to be the major regulator of platelet production. Mice deficient in either c-mpl or TPO generated by homologous recombination show a dramatic decrease in platelet counts, but other blood cell counts are normal. Because TPO treatment of myelosuppressed mice not only enhances the recovery of platelets but also accelerates erythroid recovery, we investigated the levels of myeloid and erythroid progenitor cells in TPO-or c-mpl-deficient mice. Our results show that the number of megakaryocyte, granulocyte-macrophage, erythroid, and multilineage progenitors are significantly reduced in the bone marrow, spleen, and peripheral blood of either TPO-or c-mpl-deficient mice. Administration of recombinant murine TPO to TPO-deficient mice and control littermate mice significantly increased the absolute number of myeloid, erythroid, and mixed progenitors in bone marrow and spleen. This increase was especially apparent in TPO-deficient mice where numbers were increased to a level greater than in diluent-treated control mice and approached or equaled that in the TPO-treated control mice. Moreover, TPO- administration greatly increased the number of circulating progenitors as well as platelets in both TPO-deficient and control mice. Furthermore, the megakaryocytopoietic activity of other cytokines in the absence of a functional TPO or c-mpl gene was shown both in vitro and in vivo.


Blood ◽  
1996 ◽  
Vol 88 (3) ◽  
pp. 803-808 ◽  
Author(s):  
K Carver-Moore ◽  
HE Broxmeyer ◽  
SM Luoh ◽  
S Cooper ◽  
J Peng ◽  
...  

Thrombopoietin (TPO), the ligand for the c-mpl receptor, has been shown to be the major regulator of platelet production. Mice deficient in either c-mpl or TPO generated by homologous recombination show a dramatic decrease in platelet counts, but other blood cell counts are normal. Because TPO treatment of myelosuppressed mice not only enhances the recovery of platelets but also accelerates erythroid recovery, we investigated the levels of myeloid and erythroid progenitor cells in TPO-or c-mpl-deficient mice. Our results show that the number of megakaryocyte, granulocyte-macrophage, erythroid, and multilineage progenitors are significantly reduced in the bone marrow, spleen, and peripheral blood of either TPO-or c-mpl-deficient mice. Administration of recombinant murine TPO to TPO-deficient mice and control littermate mice significantly increased the absolute number of myeloid, erythroid, and mixed progenitors in bone marrow and spleen. This increase was especially apparent in TPO-deficient mice where numbers were increased to a level greater than in diluent-treated control mice and approached or equaled that in the TPO-treated control mice. Moreover, TPO- administration greatly increased the number of circulating progenitors as well as platelets in both TPO-deficient and control mice. Furthermore, the megakaryocytopoietic activity of other cytokines in the absence of a functional TPO or c-mpl gene was shown both in vitro and in vivo.


Blood ◽  
1991 ◽  
Vol 78 (10) ◽  
pp. 2493-2497 ◽  
Author(s):  
CH Dai ◽  
SB Krantz ◽  
KM Zsebo

To understand the factors that regulate the early growth and development of immature erythroid progenitor cells, the burst-forming units-erythroid (BFU-E), it is necessary to have both highly purified target cells and a medium free of serum. When highly purified human blood BFU-E were cultured in a serum-free medium adequate for the growth of later erythroid progenitors, BFU-E would not grow even with the addition of recombinant human interleukin-3 (rIL-3), known to be essential for these cells. However, the addition of recombinant human stem cell factor (rSCF), which supports germ cell and pluripotential stem cell growth, stimulated BFU-E to grow equally well in serum-free as in serum-containing medium. Limiting dilution studies showed that rSCF acts directly on the BFU-E that do not require accessory cells for growth. Furthermore, rSCF was necessary for BFU-E development during the initial 7 days of culture, until these cells reached the stage of the late progenitors, the colony-forming units-erythroid (CFU-E). These studies indicate that early erythropoiesis is dependent on the direct action of SCF that not only affects early stem cells but is continually necessary for the further development of committed erythroid progenitor cells until the CFU-E stage of maturation.


1994 ◽  
Vol 14 (4) ◽  
pp. 2266-2277 ◽  
Author(s):  
G D Longmore ◽  
P N Pharr ◽  
H F Lodish

If the env gene of spleen focus-forming virus (SFFV) is replaced by a cDNA encoding a constitutively active form of the erythropoietin receptor, EPO-R(R129C), the resultant recombinant virus, SFFVcEPO-R, induces transient thrombocytosis and erythrocytosis in infected mice. Clonogenic progenitor cell assays of cells from the bone marrow and spleens of these infected mice suggest that EPO-R(R129C) can stimulate proliferation of committed megakaryocytic and erythroid progenitors as well as nonerythroid multipotent progenitors. From the spleens of SFFVcEPO-R-infected mice, eight multiphenotypic immortal cell lines were isolated and characterized. These included primitive erythroid, lymphoid, and monocytic cells. Some expressed proteins characteristic of more than one lineage. All cell lines resulting from SFFVcEPO-R infection contained a mutant form of the p53 gene. However, in contrast to infection by SFFV, activation of PU.1 gene expression, by retroviral integration, was not observed. One cell line had integrated a provirus upstream of the fli-1 gene, in a location typically seen in erythroleukemic cells generated by Friend murine leukemia virus infection. This event led to increased expression of fli-1 in this cell line. Thus, infection by SFFVcEPO-R can induce proliferation and lead to transformation of nonerythroid as well as very immature erythroid progenitor cells. The sites of proviral integration in clonal cell lines are distinct from those in SFFV-derived lines.


Blood ◽  
1996 ◽  
Vol 88 (5) ◽  
pp. 1576-1582 ◽  
Author(s):  
M Silva ◽  
D Grillot ◽  
A Benito ◽  
C Richard ◽  
G Nunez ◽  
...  

Abstract Erythropoietin (Epo), the hormone that is the principal regulator of red blood cell production, interacts with high-affinity receptors on the surface of erythroid progenitor cells and maintains their survival. Epo has been shown to promote cell viability by repressing apoptosis; however, the molecular mechanism involved is unclear. In the present studies we have examined whether Epo acts as a survival factor through the regulation of the bcl-2 family of apoptosis-regulatory genes. We addressed this issue in HCD-57, a murine erythroid progenitor cell line that requires Epo for proliferation and survival. When HCD-57 cells were cultured in the absence of Epo, Bcl-2 and Bcl-XL but not Bax were downregulated, and the cells underwent apoptotic cell death. HCD-57 cells infected with a retroviral vector encoding human Bcl-XL or Bcl-2 rapidly stopped proliferating but remained viable in the absence of Epo. Furthermore, endogenous levels of bcl-2 and bcl-XL were downregulated after Epo withdrawal in HCD-57 cells that remained viable through ectopic expression of human Bcl-XL, further indicating that Epo specifically maintains the expression of bcl-2 and bcl-XL. We also show that HCD-57 rescued from apoptosis by ectopic expression of Bcl-XL can undergo erythroid differentiation in the absence of Epo, demonstrating that a survival signal but not Epo itself is necessary for erythroid differentiation of HCD-57 progenitor cells. Thus, we propose a model whereby Epo functions as a survival factor by repressing apoptosis through Bcl-XL and Bcl-2 during proliferation and differentiation of erythroid progenitors.


Blood ◽  
1997 ◽  
Vol 90 (2) ◽  
pp. 651-657 ◽  
Author(s):  
Xingwei Sui ◽  
Sanford B. Krantz ◽  
Zhizhuang Zhao

Abstract Polycythemia vera (PV) is a clonal hematologic disease characterized by hyperplasia of the three major bone marrow lineages. PV erythroid progenitor cells display hypersensitivity to several growth factors, which might be caused by an abnormality of tyrosine phosphorylation. In the present study, we have investigated protein tyrosine phosphatase (PTP) activity in highly purified erythroid progenitor cells and found that the total PTP activity in the PV cells was twofold to threefold higher than that in normal cells. Protein separation on anion-exchange and gel-filtration columns showed that the increased activity was due to a major PTP eluted at approximately 170 kD. This enzyme was sensitive to PTP inhibitors and it did not cross-react with antibodies to SHP-1, SHP-2, or CD45. Subcellular fractionation showed that the PTP localized with the membrane fraction, where its activity was increased by threefold in PV erythroid progenitors when compared with normal cells. As the erythroid progenitors progressively matured, activity of the PTP declined rapidly in the normal cells but at a much slower rate in the PV cells. These studies suggest that a potentially novel membrane or membrane-associated PTP, representing a major PTP activity, may have an important role in proliferation and/or survival of human erythroid progenitors and that its hyperactivation in PV erythroid progenitors might be responsible for the increased erythropoiesis in PV patients.


Blood ◽  
2004 ◽  
Vol 103 (10) ◽  
pp. 3615-3623 ◽  
Author(s):  
Jonathan Back ◽  
Andrée Dierich ◽  
Corinne Bronn ◽  
Philippe Kastner ◽  
Susan Chan

Abstract PU.1 is a hematopoietic-specific transcriptional activator that is absolutely required for the differentiation of B lymphocytes and myeloid-lineage cells. Although PU.1 is also expressed by early erythroid progenitor cells, its role in erythropoiesis, if any, is unknown. To investigate the relevance of PU.1 in erythropoiesis, we produced a line of PU.1-deficient mice carrying a green fluorescent protein reporter at this locus. We report here that PU.1 is tightly regulated during differentiation—it is expressed at low levels in erythroid progenitor cells and down-regulated upon terminal differentiation. Strikingly, PU.1-deficient fetal erythroid progenitors lose their self-renewal capacity and undergo proliferation arrest, premature differentiation, and apoptosis. In adult mice lacking one PU.1 allele, similar defects are detected following stress-induced erythropoiesis. These studies identify PU.1 as a novel and critical regulator of erythropoiesis and highlight the versatility of this transcription factor in promoting or preventing differentiation depending on the hematopoietic lineage.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1260-1260
Author(s):  
Melissa M. Rhodes ◽  
Prapaporn Kopsombut ◽  
Maurice Bondurant ◽  
James O. Price ◽  
Mark J. Koury

Abstract Bcl-x is a protein in the outer mitochondrial membrane. A member of the Bcl-2 family, Bcl-x protects developing erythroid cells from apoptosis. The exact stage of erythroid development at which Bcl-x exerts its anti-apoptotic effect is not known, but induction of Bcl-x has been proposed to be the mediator of erythropoietin’s (EPO) anti-apoptotic effect in erythroid differentiation. EPO is the principal trophic hormone that controls red blood cell production by regulating apoptosis of erythroid progenitor cells at the CFU-E and early erythroblast stages. Bcl-x has also been reported to be necessary for heme synthesis. In mice, Bcl-x deficiency is embryonically lethal; when Bcl-x deficiency is acquired postnatally by conditional knockout technology, it is associated with splenomegaly, thrombocytopenia, and a profound anemia that is thought to be hemolytic in origin. Objectives: 1)To characterize the defect of erythroid differentiation in conditional Bcl-x −/− mice. 2)To determine whether Bcl-x is the mediator of EPO’s anti-apoptotic action. 3)To determine whether Bcl-x is necessary for heme synthesis. Methods: Phlebotomized or unbled littermate controls and anemic adult Bcl-x −/− mice obtained by cre-lox conditional knockout were bled, sacrificed, and splenectomized. Purified populations of splenic erythroblasts were isolated by sedimentation at unit gravity, cultured with or without EPO, and harvested at 0, 8, 20, 32, and 44 hours for cell counts, cytospin preparations for morphology, flow cytometry analyses for apoptosis (TUNEL) and cell cycle phases, and 59FeCl3 incorporation into heme. Results: Compared to littermate controls, Bcl-x −/− mice were severely anemic (Hgb 2.8 g/dL vs 15.4 g/dL in unbled controls and 7.2 g/dL in bled controls), thrombocytopenic (platelets 23x103/microL vs 905x103/microL in unbled controls and 984x103/microL in bled controls), and reticulocytopenic (82.8x103/microL vs 281x103/microL in unbled controls and 1410x103/microL in bled controls), while WBCs were unaffected. Expanded erythropoiesis led to massive splenomegaly (spleens =4.3gm vs 0.1gm in unbled controls and 0.3gm in bled controls). After 44 hours of culture with EPO, purified erythroblasts from bled controls proliferated 4-fold and differentiated such that the majority enucleated, producing 200–250 reticulocytes per 100 erythroblasts plated, whereas Bcl-x −/− erythroblast numbers doubled during the first 20 hours in culture, but the large majority died by apoptosis between 20 and 44 hours, producing only 9–12 reticulocytes per 100 erythroblasts plated. Bcl-x −/− erythroblast apoptosis occurred after the initiation of heme synthesis and proportionally in all phases of cell cycle. Compared to culture with EPO, Bcl-x −/− erythroblasts cultured without EPO underwent increased apoptosis at earlier times of culture-- at 8 hours (45% vs 29%), 20 hours (71% vs 42%) and 32 hours (83% vs 57%). Conclusions: 1)Bcl-x is required for the survival and differentiation of the late-stage erythroblasts in all phases of cell cycle. Thus, Bcl-x deficiency results in ineffective erythropoiesis rather than hemolytic anemia. 2)Bcl-x is not required for heme synthesis, but has its anti-apoptotic effect during the stage of hemoglobin synthesis. 3)Bcl-x does not mediate EPO’s anti-apoptotic effect in early-stage erythroblasts.


Sign in / Sign up

Export Citation Format

Share Document