In vitro proliferation and differentiation of human CD34+ cells from peripheral blood into mature red blood cells with two different cell culture systems

Transfusion ◽  
2008 ◽  
Vol 48 (6) ◽  
pp. 1122-1132 ◽  
Author(s):  
Isabel Dorn ◽  
Pamela Lazar-Karsten ◽  
Stefanie Boie ◽  
Julika Ribbat ◽  
Dirk Hartwig ◽  
...  
Blood ◽  
2002 ◽  
Vol 99 (5) ◽  
pp. 1594-1601 ◽  
Author(s):  
Yann-Erick Claessens ◽  
Didier Bouscary ◽  
Jean-Michel Dupont ◽  
Françoise Picard ◽  
Josiane Melle ◽  
...  

Erythropoiesis results from the proliferation and differentiation of pluripotent stem cells into immature erythroid progenitors (ie, erythroid burst-forming units (BFU-Es), whose growth, survival, and terminal differentiation depends on erythropoietin (Epo). Ineffective erythropoiesis is a common feature of myelodysplastic syndromes (MDS). We used a 2-step liquid-culture procedure to study erythropoiesis in MDS. CD34+ cells from the marrow of patients with MDS were cultured for 10 days in serum-containing medium with Epo, stem cell factor, insulinlike growth factor 1, and steroid hormones until they reached the proerythroblast stage. The cells were then placed in medium containing Epo and insulin for terminal erythroid differentiation. Numbers of both MDS and normal control cells increased 103fold by day 15. However, in semisolid culture, cells from patients with refractory anemia (RA) with ringed sideroblasts and RA or RA with excess of blasts produced significantly fewer BFU-Es than cells from controls. Fluorescence in situ hybridization analysis of interphase nuclei from patients with chromosomal defects indicated that abnormal clones were expanded in vitro. Epo-signaling pathways (STAT5, Akt, and ERK 1/2) were normally activated in MDS erythroid progenitors. In contrast, apoptosis was significantly increased in MDS cells once they differentiated, whereas it remained low in normal cells. Fas was overexpressed on freshly isolated MDS CD34+ cells and on MDS erythroid cells throughout the culture. Apoptosis coincided with overproduction of Fas ligand during the differentiation stage and was inhibited by Fas-Fc chimeric protein. Thus, MDS CD34+-derived erythroid progenitors proliferated normally in our 2-step liquid culture with Epo but underwent abnormal Fas-dependent apoptosis during differentiation that could be responsible for the impaired erythropoiesis.


2019 ◽  
Author(s):  
Shouping Zhang ◽  
Emmanuel N Olivier ◽  
Zi Yan ◽  
Sandra Suzuka ◽  
Karl Roberts ◽  
...  

AbstractMany methods have been developed to produce red blood cellsin vitrobut translational applications have been hampered by the high cost of production. We have developed R6, a chemically-defined, albumin-free, low-transferrin culture medium, and MNC-RED, a protocol to differentiate peripheral blood mononuclear cells into enucleated erythroid cells that does not require any albumin or any animal components. Erythropoiesis requires large amounts of iron for hemoglobin synthesis. In all existing protocols, these large iron needs are met by increasing the concentration of holo-transferrin. This is necessary because transferrin recycling does not take place in existing erythroid culture conditions. In the R6 medium, iron is provided to the differentiating erythroblasts by small amounts of recombinant transferrin supplemented with FeIII-EDTA, an iron chelator that allows transferrin recycling to take place in cell culture. As a result of the absence of albumin and the use of low amounts of transferrin, the production of cultured red blood cells using the MNC-RED protocol is much less expensive than with existing protocols. The MNC-RED protocol should therefore help make the many translational applications of cultured RBCs economically more feasible.HighlightsWe have developed R6, a chemically-defined, albumin-free low-transferrin culture medium, and MNC-RED, a protocol to differentiate peripheral blood mononuclear cells into enucleated erythroid ER6 is suitable for red blood cell culture despite the low transferrin amounts because of the presence of FeIII-EDTA, an iron chelator that allows transferrin recycling to take place in cell culture.The MNC-RED protocol should help make the many translational applications of cultured RBCs more economically feasible.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2315-2315
Author(s):  
Tyler A Couch ◽  
Zachary C. Murphy ◽  
Michael Getman ◽  
Ryo Kurita ◽  
Yukio Nakamura ◽  
...  

Abstract There is a constant need for red blood cells for transfusion therapy in the treatment of anemias and acute injury. As all blood products for transfusion come from donors, there are concerns over shortages and safety. Furthermore, many patients with transfusion-dependent anemias risk alloiumminization. The in vitro production of red blood cells would address these problems, especially as they can be genetically engineered to prevent alloimmunization. Numerous erythroid culture systems now exist for the in vitro production of red blood cells. Hematopoietic stem and progenitor cells (HSPCs) obtained from umbilical cord or peripheral blood can be differentiated into erythrocytes, however, they are limited in expansion. While umbilical cord HSPCs have greater expandability than peripheral blood, the resulting erythrocytes contain fetal globins. Pluripotent stem cells can also be used as a starting source, however only a small percentage of the cells can be differentiated into erythroblasts which also suffer from low enucleation rates. Presently, the cost of in vitro production of a unit of red cells is greater than an order of magnitude higher than obtaining it from a donor largely due to the medium and cytokine costs (Timmins & Nielsen, Trends Biotechnol, 2009). A relatively new approach of immortalizing early erythroblasts allowing unlimited expansion as well as terminal maturation and enucleation shows great therapeutic promise (Kurita et al., PLoS One, 2013; Huang et al., Mol Ther, 2014; Trakarnsanga et al., Nat Commun, 2017). However, these immortalized erythroblasts are still reliant on two costly cytokines: stem cell factor (SCF) and erythropoietin (Epo). Mutations in exon 17 of the receptor tyrosine kinase gene KIT are frequently seen in acute myeloid leukemias, gastrointestinal stromal tumors, and mast cells leading to mastocytosis. These mutations cause the c-Kit protein to spontaneously activate and transduce signal in the absence of SCF (Kit-ligand). To generate an SCF-independent HUDEP-2 cell line (Kurita et al., PLoS One, 2013), we used CRISPR/Cas9 to introduce missense and frameshifting mutations within the vicinity of Asp816 in exon 17 of the KIT gene. The resulting monoclonal cell lines were selected for by removing SCF from the expansion medium and were subsequently named KIT-CAT (KIT with Constitutively Activating Transformation). To better understand what KIT mutations allowed or impaired terminal maturation, monoclonal cell lines were genotyped by Sanger sequencing. Three cell lines with unique genotypes were chosen for further analysis. All three KIT-CAT lines had a shorter doubling time compared to HUDEP-2 cells (16.7 vs 18.9 hrs, p=0.020) and were no longer dependent on SCF or Epo. However, two of the three KIT-CAT lines showed more robust proliferation with Epo in the expansion medium. The addition of SCF to the medium caused no increase in c-Kit activation by Western blotting for phosphorylation at Tyr703. Furthermore, the low molecular weight and immature form of c-Kit is also phosphorylated in KIT-CAT cells, but not HUDEP-2 cells, indicating c-Kit activation occurs before trafficking to the cell membrane where SCF would bind (Tabone-Eglinger et al., Clin Cancer Res, 2008). Key features of erythroblast maturation are the decrease in cell and nuclear size which can be measured using imaging flow cytometry (McGrath et al., Methods, 2017). While in expansion phase, all 3 cell lines were larger in cell and nuclear area compared to the parental HUDEP-2 line. By day 6 of maturation, all three cell lines had statistically significant decreases in cell and nuclear size indicating maturation. By day 13 of culture, Wright-Giemsa staining showed that the majority of the cells were orthochromatic erythroblasts or enucleate reticulocytes. Reducing cell culture costs is needed for in vitro manufacturing of red blood cells to be economically feasible. These results show that a c-Kit activating mutations in human erythroblasts removes the cost of SCF and reduces the cost of Epo while still allowing for terminal maturation and enucleation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4748-4748
Author(s):  
Daniela Boehm ◽  
Mohamed Al-Rubeai ◽  
William G Murphy

Abstract Erythropoiesis is one of the body’s most productive cell production processes yielding 2×1011 new red cells from hematopoietic stem cells (HSCs) of the bone marrow every day. Intensive research has focused on mimicking this process ex vivo through application of various growth factor combinations or co-culture with stromal cells. To develop a scalable and reproducible system for large scale production of red blood cells we have investigated in vitro erythropoiesis of peripheral blood derived CD34+ cells with primary focus on the impact of the microenvironment on the process. The influence of cultivation conditions on expansion of erythroid progenitor cells and their terminal differentiation to mature red blood cells were studied in stroma-free liquid culture supplemented with stem cell factor (SCF), interleukin-3 (IL-3) and erythropoietin (EPO). Peripheral blood derived CD34+ cells were expanded by more than 105 fold over a 3 week period. This degree of expansion has only been achieved previously for CD34+ cells derived from more potent stem cell sources such as cord blood, bone marrow and G-CSF mobilized peripheral blood (Giarratana et al, Nat Biotechnol 2005). The natural environment of human erythropoiesis, the bone marrow, is a very crowded milieu where hematopoietic precursors and other cells are packed in close proximity. Cell crowdedness was found to have significant influences on ex vivo erythropoiesis. Cell density per surface area rather than cell concentration per media volume determined cell expansion during exponential growth where more crowded cells showed reduced overall expansion. In cultures inoculated at 4×105 cells/ml (2.1×105 cells/cm2) increasing cell density per area (i.e. decreasing surface area to volume ratio) 4fold (to 8.4×105 cells/cm2) resulted in 35±12% reduction of total expansion (p<0.05, unpaired Student’s t-test). While 4fold increase of cell density in cultures seeded at 1×106 cells/ml (from 5.3×105 cells/cm2 to 2.1×106 cells/cm2) reduced overall expansion by 51±9% (p<0.01). In late stage erythropoiesis, however, when cells had become arrested in G1 and no longer proliferated, cell density was seen to enhance cell viability. Dilution series of late stage erythroblasts showed that although cell viability gradually decreased over a 14 day cultivation period the decreasing rate was lower in cells cultivated at higher density as shown in the Figure. Enhanced viability in crowded culture conditions could reflect the cells’ dependency on direct cell-cell interactions as found in the marrow environment. Cultures grown to high cell densities of 2–3×106 cells/cm2 showed higher maturation efficiency than previously obtained in this cultivation set-up with more than 80% of cells being CD71-/GpA+. Enucleation yields of up to 45% were achieved indicating a significant amount of terminal maturation to red blood cells. Efficient maturation and particularly enucleation have in many cases been found to be dependent on or improved by interactions with feeder cells or macrophages (Fujimi et al, Int J Hematol 2008). Keeping erythroid cells at high densities during late stages of erythropoiesis possibly helps to mimic their in vivo environment, thus allowing for better survival and efficient terminal maturation without the need for co-culture with other cells. Figure Figure


1997 ◽  
Vol 185 (1) ◽  
pp. 111-120 ◽  
Author(s):  
A. Aiuti ◽  
I.J. Webb ◽  
C. Bleul ◽  
T. Springer ◽  
J.C. Gutierrez-Ramos

Hematopoietic progenitor cells migrate in vitro and in vivo towards a gradient of the chemotactic factor stromal cell-derived factor-1 (SDF-1) produced by stromal cells. This is the first chemoattractant reported for human CD34+ progenitor cells. Concentrations of SDF-1 that elicit chemotaxis also induce a transient elevation of cytoplasmic calcium in CD34+ cells. SDF-1-induced chemotaxis is inhibited by pertussis toxin, suggesting that its signaling in CD34+ cells is mediated by seven transmembrane receptors coupled to Gi proteins. CD34+ cells migrating to SDF-1 include cells with a more primitive (CD34+/CD38− or CD34+/DR−) phenotype as well as CD34+ cells phenotypically committed to the erythroid, lymphoid and myeloid lineages, including functional BFU-E, CFU-GM, and CFU-MIX progenitors. Chemotaxis of CD34+ cells in response to SDF-1 is increased by IL-3 in vitro and is lower in CD34+ progenitors from peripheral blood than in CD34+ progenitors from bone marrow, suggesting that an altered response to SDF-1 may be associated with CD34 progenitor mobilization.


2015 ◽  
Vol 37 (1) ◽  
pp. 70-72 ◽  
Author(s):  
I O Sviezhentseva ◽  
T P Perekhrestenko ◽  
D I Bilko ◽  
A I Gordienko ◽  
M V Diachenko ◽  
...  

Introduction: It is believed that the reason of the leukemic clone cell resistance to treatment with tyrosine kinase inhibitors during chronic myeloid leukemia (CML) is mutations in the genome of an early bone marrow progenitor cells that are CD34-positive. Such cells, regardless of treatment, acquire ability to proliferation and differentiation. This leads to the re-expansion of the CD34+ cells. Aim: to determine the CD34 antigen expression in bone marrow and peripheral blood cells in CML patients with different response to imatinib therapy using the results of hematopoietic cells culturing and the data of flow cytometry. Methods: Bone marrow aspirate from 39 patients who were treated with imatinib was studied with cytogenetic, flow cytometry and culture methods in vitro. Results: In patients with an optimal response to imatinib therapy the number of colonies was 1.8 times lower than the number of those in the group of patients with a suboptimal response to therapy. In turn, in patients with failure of imatinib therapy the number of colonies was the highest and was 2.1 times higher than the patients with optimal response. The results of cytometric studies have shown that the number of CD34+ cells in bone marrow was significantly higher compared to the number of CD34+ cells in peripheral blood cells and increased with the acquisition of leukemic cells the resistance to imatinib. There was a direct correlation between the number of colonies and clusters in semisolid agar in vitro and the number of CD34+ cells in the bone marrow of patients. Conclusions: The correlation between the number of CD34+ cells and the number of cell aggregates in semisolid agar in vitro indicates the prognostic value of the method for determining CD34+ cells in the patient bone marrow. The parallel increase of their number in the peripheral blood will allow developing express methods for the detection of individual patient response to imatinib therapy.


1996 ◽  
Vol 15 (1) ◽  
pp. 26-29 ◽  
Author(s):  
C. Gachet ◽  
R. Ennamany ◽  
O. Kretz ◽  
P. Ohlmann ◽  
C. Krause ◽  
...  

Bolesatine is a toxic glycoprotein isolated from the mushroom Boletus satanas Lenz, which has been shown to inhibit protein synthesis in cell-free systems and cell culture. It is toxic to rodents, the LD50% 24 h being 1 mg kg-1 (i.p.) and 0.15 mg kg-1 (i.v) in the rat in which it induces hepatic blood stasis. Bolesatine possesses lectinic properties with in parti cular a sugar binding site for D-galactose and mitogenic activity toward lymphocytes. Tested for cell agglutination on red blood cells and platelets, bolesatine agglutinates both human and rat platelets from threshold concentrations of 30 and 300 nM respectively. EDTA and PGI2 (aggregation inhibitors) do not decrease the agglutination induced by bolesatine, indicating that the process does not involve platelet activation. In contrast, fibrinogen decreases platelet agglutination in duced by bolesatine, most likely by masking the binding sites on platelets or by interacting with the toxin. Bolesatine agglutinates all red blood cells without any blood group specificity in the concentration range of 20 to 40 nM. This haemagglutination cannot be prevented by sugars, including D-galactose at a concentration of 0.5 M.


Sign in / Sign up

Export Citation Format

Share Document