scholarly journals Humanized Culture Medium for Clinical Expansion of Human Erythroblasts

2010 ◽  
Vol 19 (4) ◽  
pp. 453-469 ◽  
Author(s):  
Giovanni Migliaccio ◽  
Massimo Sanchez ◽  
Francesca Masiello ◽  
Valentina Tirelli ◽  
Lilian Varricchio ◽  
...  

Ex vivo-generated erythroblasts represent alternative transfusion products. However, inclusion of bovine components in media used for their growth precludes clinical use, highlighting the importance of developing culture media based on pharmaceutical grade reagents. In addition, because adult blood generates ex vivo lower numbers of erythroblasts than cord blood, cord blood has been proposed as the source of choice for ex vivo erythroblast production. To clarify the potential of adult blood to generate erythroblasts ex vivo, experiments were designed to identify growth factors [stem cell factor (SCF), interleukin-3 (IL-3), erythropoietin (EPO), and/or thrombopoietin (TPO)] and the optimal concentration and addition schedule of hormones (dexamethasone and estradiol) sustaining maximal erythroid amplification from adult blood mononuclear cells (MNC) using media with serum previously defined as human erythroid massive amplification culture (HEMAser). Adult MNC stimulated with SCF and IL-3 in combination with EPO generated a 6–12-fold increase in erythroid cells while TPO was ineffective. Dexamethasone and estradiol (both at 10−6 M) exerted partially overlapping but nonredundant functions. Dexamethasone was indispensable in the first 10 days of culture while estradiol was required from day 10 on. The growth factor and hormone combinations identified in HEMAser were then used to formulate a media composed of dialyzed pharmaceutical grade human albumin, human albumin-lipid liposomes, and iron-saturated recombinant human tranferrin (HEMAdef). HEMAdef sustained erythroid amplification as efficiently as HEMAser for cord blood MNC and 10-fold higher than HEMAser for adult blood MNC. In fact, the numbers of erythroblasts generated in HEMAdef by adult MNC were similar to those generated by cord blood MNC. In conclusion, this study identifies growth factors, hormone combinations, and human protein-based media that allow similar levels of ex vivo erythroid expansion from adult and cord blood MNC, paving the way to evaluate adult blood as a source of ex vivo-expanded erythroblasts for transfusion.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 642-642
Author(s):  
Giovanni Migliaccio ◽  
Massimo Sanchez ◽  
Francesca Masiello ◽  
Valentina Tirelli ◽  
Lilian Varricchio ◽  
...  

Abstract Abstract 642 Ex-vivo generated erythroblasts (EBs) represent alternative transfusion products. Adult blood (AB) contains numbers of progenitor cells comparable to those present in cord blood (CB) (106 vs 1.8×106 CD34pos cells in average AB and CB donations) but generates lower numbers of erythroblasts (EBs) (∼4.8×108 vs 6.6×1010, respectively) and, in spite of its numerous advantages, is not considered suitable for ex-vivo EB production. To assess the potential of AB to generate EBs ex-vivo, the growth factors [stem cell factor (SCF), interleukin-3 (IL-3) and erythropoietin (EPO)] and optimal concentration and addition schedule of dexamethasone (DXM) and estradiol (ES) sustaining maximal EB amplification from AB mononuclear cells (MNC) were defined using media with serum previously defined as human erythroid massive amplification culture (HEMAser). Adult MNC stimulated with SCF and IL-3 in combination with EPO generated low numbers (fold increase ∼2) of EBs at all stages of maturation. Concentration response studies conducted on MNC from 10 different donors, indicated that the further addition to the cultures of DXM and ES (both at 10-6 M) increased (∼6-12-fold) the numbers of EBs generated. Delayed addition and withdrawal experiments indicated that DXM and ES exerted partially overlapping but non-redundant functions. DXM was indispensable to achieve maximal amplification in the first 10 days of culture while ES was required from day 10 on. To determine if variability in glucocorticoid receptor (GR) expression might affect ex vivo generation of EBs, expression of αa and γ GR isoforms (αaGR and γGR) by EBs from 10 AB and 5 CB was investigated. While EBs from all donors expressed αaGR, γGR was not expressed by EBs obtained from CB and from AB that generated high numbers of EBs ex vivo, suggesting that activation of γGR in EBs is ontogenetically activated in a subset of AB and may predicts poor expansion. Ex vivo produced EBs are megaloblastic (30 to 50 μm). EPO decreased their size from 40.1±1.4 to 11.6±0.3 μm by 96 h (p<0.01). Although still macrocytic (adult normocytic red cells are 8 μm), these cells are smaller than fetal red cells (12.5 μm) and therefore suitable for clinical use. Inclusion of bovine components in HEMAser precludes its use for clinical purposes. Therefore, optimal growth factor and hormone combinations identified in HEMAser were used to formulate a medium composed of pharmaceutical grade human albumin, human albumin-based-lipid liposomes and iron-saturated recombinant human-tranferrin (HEMAdef). HEMAdef sustained EB amplification as efficiently as HEMAser from CB MNC and 10-fold higher than HEMAser from AB MNC. Moreover, the numbers of EBs generated in HEMAdef by adult MNC were similar to those generated by CB MNC (750×106 vs 500×106 per 106 MNC from AB and CB, respectively). Assuming that MNC contain 102-103 EB progenitors (CD34pos cells represent 0.1% of MNC and erythroid progenitors represent 10% of CD34pos cells), it was calculated that the generation of 750×106 EBs from the progenitors present in 106 adult MNC required 19-23 divisions, a number below the theoretical Hayflick's limit for somatic cell divisions of 35. These results indicate that at least a subset of AB donors is suitable to produce ex-vivo erythroid cells for transfusion and that it should be possible, by optimizing HEMAdef components, to further increase the number of EBs that can be generated ex-vivo from AB. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3462-3462
Author(s):  
Varda R Deutsch ◽  
Einav Hubel ◽  
Sigi Kay ◽  
Tal Ohayon ◽  
Ben-Zion Katz ◽  
...  

Abstract While umbilical cord blood provides an important source of hematopoietic stem/progenitor cells (HSPC) for allogeneic transplantation in children, its use in adults is limited by the inadequate number of primitive stem cells and megakaryocyte progenitors (Mk-p) in single or even double CB units resulting in prolonged thrombocytopenia. Thrombopoietin treatment is not effective in these patients due to the paucity of target progenitors and patients require multiple platelet transfusions until the long-term engrafting cells can support thrombopoiesis, thus new modalities to increase progenitor cell dose are needed. A new transplantation strategy could involve the infusion of ex vivo-generated Mk-p together with unmanipulated single or double CB units. While CB CD34+ cells can be expanded by several reported methods, these rare cells cannot be sacrificed from the CB units due to their limited number. We propose a novel ex-vivo strategy to facilitate HSPC and Mk-p expansion from mononuclear cells (MNC) of a small aliquot of CB using conditions that mimic the hematopoietic niche, in short term cultures. Fibronectin (FN) was considered to be a prime candidate to support proliferation because it is a major extracellular matrix (ECM) component of all bone marrow hematopoietic microenvironments which is known to enhance viability and proliferation of HSPC. Other growth stimulators added were thrombopoietin (r-hu-TPO), the major physiological stimulator of MK and the synthetic hematopoietic stress peptide ARP derived from acetylcholinesterase, shown to increase transplantable Mk-p and produce human platelets in NOD/SCID mice (Pick et al, Blood 2006, Grisaru et al, J Imm 2006). High definition flow cytometry enabled assessing expansion of the SSClow/CD34high HSPC, and the SSClow/CD45dim/neg/CD41high Mk-p, and their subpopulations on day 0 and 10 of culture. True MK expansion was assessed by gating out of granulocyte and monocytes, which acquire CD41+ adherent platelets in culture. FN alone increased viability and expansion of HSPC by 6.9 fold and MK-p by 4-fold, while r-hu-TPO alone enhanced Mk-p proliferation with an average expansion of 8.3-fold in agreement with its known activity. Combining FN with r-hu-TPO produced a 25-fold increase in the number of MK-p while adding ARP to FN and r-hu-TPO was even more powerful, doubling the number of cells with a highly significant average expansion of 59-fold (p < 0.001). To define the progenitor subpopulations that contributed to Mk-p proliferation with FN, r-hu-TPO and ARP, we further analyzed the resulting subsets of MK-p cells, which also expressed either CD34, or the early myeloid marker CD33. The CD41high/CD34high population was increased by 4 fold, while the CD41high/CD33+ Mk-p, a subset with properties similar to clonogenic GEMM progenitors that could provide both myeloid and megakaryocytic cells post-transplant, were stimulated 30–50 fold. This notion is confirmed by the stimulation of CFU-MK and CFU-GEMM obtained under these conditions. Considering that expansion of MK-p requires proliferation of the HSPC precursor, we examined the proliferation of CD34+ progenitor cells and their subpopulations; CD34high/CD33+ or CD34high/CD41low uncommitted HSPC and CD41 high committed Mk subpopulations. The addition of FN alone stimulated CD34+ HSPC expansion by 6.9-fold (p < 0.05). All cultures that contained the ARP peptide maintained a high proliferation capacity, confirming that ARP protects and drives CD34+HSPC and early myeloid cell proliferation (Deutsch et al Exp Hem 2002). The addition of r-hu-TPO and ARP to FN produced a synergistic proliferative effect on the CD34+/CD41low HSPC stimulating a dramatic 440 fold increase of these uncommitted cells. These data support the notion that FN is protective and plays an essential role in enabling HSPC and MK-p expansion driven by r-hu-TPO and ARP. These conditions also supported MK maturation, as measured by increased high ploidy cells and elevated expression of GPIIb/IIIa detected by quantitative real time PCR. We demonstrate that expansion of both very early myeloid and Mk-p from a small fraction of the CB unit in short term cultures under conditions that mimic the hematopoietic niche is feasible, easy to perform and can comply with GTP requirements. This approach may lead to the development of more effective cell therapy modalities to facilitate myelopoiesis and platelet production following CBT.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Sara Bucar ◽  
André Dargen de Matos Branco ◽  
Márcia F. Mata ◽  
João Coutinho Milhano ◽  
Íris Caramalho ◽  
...  

Abstract Background Umbilical cord blood (UCB) is a clinically relevant alternative source of hematopoietic stem/progenitor cells (HSPC). To overcome the low cell number per UCB unit, ex vivo expansion of UCB HSPC in co-culture with mesenchymal stromal cells (MSC) has been established. Bone marrow (BM)-derived MSC have been the standard choice, but the use of MSC from alternative sources, less invasive and discardable, could ease clinical translation of an expanded CD34+ cell product. Here, we compare the capacity of BM-, umbilical cord matrix (UCM)-, and adipose tissue (AT)-derived MSC, expanded with/without xenogeneic components, to expand/maintain UCB CD34+-enriched cells ex vivo. Methods UCB CD34+-enriched cells were isolated from cryopreserved mononuclear cells and cultured for 7 days over an established feeder layer (FL) of BM-, UCM-, or AT-derived MSC, previously expanded using fetal bovine serum (FBS) or fibrinogen-depleted human platelet lysate (HPL) supplemented medium. UCB cells were cultured in serum-free medium supplemented with SCF/TPO/FLT3-L/bFGF. Fold increase in total nucleated cells (TNC) as well as immunophenotype and clonogenic potential (cobblestone area-forming cells and colony-forming unit assays) of the expanded hematopoietic cells were assessed. Results MSC from all sources effectively supported UCB HSPC expansion/maintenance ex vivo, with expansion factors (in TNC) superior to 50x, 70x, and 80x in UCM-, BM-, and AT-derived MSC co-cultures, respectively. Specifically, AT-derived MSC co-culture resulted in expanded cells with similar phenotypic profile compared to BM-derived MSC, but resulting in higher total cell numbers. Importantly, a subpopulation of more primitive cells (CD34+CD90+) was maintained in all co-cultures. In addition, the presence of a MSC FL was essential to maintain and expand a subpopulation of progenitor T cells (CD34+CD7+). The use of HPL to expand MSC prior to co-culture establishment did not influence the expansion potential of UCB cells. Conclusions AT represents a promising alternative to BM as a source of MSC for co-culture protocols to expand/maintain HSPC ex vivo. On the other hand, UCM-derived MSC demonstrated inferior hematopoietic supportive capacity compared to MSC from adult tissues. Despite HPL being considered an alternative to FBS for clinical-scale manufacturing of MSC, further studies are needed to determine its impact on the hematopoietic supportive capacity of these cells.


2014 ◽  
Vol 26 (1) ◽  
pp. 154 ◽  
Author(s):  
D. Moreno ◽  
A. Neira ◽  
L. Dubreil ◽  
L. Liegeois ◽  
S. Destrumelle ◽  
...  

In the majority of media for embryo culture, 2 of typical components used are FCS or BSA; however, the presence of FCS in the culture medium has been shown to have a negative effect on embryo quality and the use of animal-derived proteins in culture media increases the risks of disease transmission through in vitro embryo production. The aim of this study was to develop an in vitro embryo culture medium free from FCS and BSA, but with the addition of various growth factors and cytokines (GF-CYK: IGF-I, IGF-II, bFGF, LIF, GM-CSF) 50 ng mL–1 and (TGF-β1) 100 ng mL–1 supplemented with hyaluronan (HA) and recombinant albumin (RA). Bovine oocytes (n = 1043, 6 replicates) from abattoir ovaries were matured in TCM-199 medium with 60 μg mL–1 penicillin, 60 μg mL–1 streptomycin, and 10 ng mL–1 EGF for 24 h at 39°C and 5% CO2 in humidified air. Afterward, the oocytes were fertilized in IVF-TALP medium with 6 mg mL–1 fatty acid-free BSA and 1.7 IU mL–1 heparin for 18 h under the same conditions. After fertilization, presumptive zygotes were divided into two groups and cultured in 30 μL droplets of SOF supplemented with (1) 0.4% BSA + 5 μg mL–1 insulin, 5 μg mL–1 transferrin, and 5 ng mL–1 selenium (ITS) as a control; or (2) GF-CYK + 0.5 mg mL–1 HA + 0.15% RA (M1). Droplets were preserved under mineral oil in a humidified atmosphere of 5% CO2, 5% O2, and 90% N2 at 39°C. Blastocyst development and blastocyst diameter was observed at 7 and 8 days post-fertilization (dpf). Developmental and diameter data were analysed using the Wilcoxon test by using R software. The blastocyst rates were not significantly different between the control and M1 medium: at 7 dpf (22.9% ± 4.8 and 30.2% ± 3.0), and at 8 dpf (29.6% ± 5.1 and 37.4% ± 2.0 respectively; P > 0.05). The blastocyst diameter obtained with the M1 medium was significantly greater (P < 0.05) than that of the control at 7 dpf (173.3 μm ± 4.9 and 157.2 μm ± 4.1, respectively); however, no significant differences were observed at 8 dpf (190.3 μm ± 5.2 and 179.7 μm ± 5.3, respectively). In conclusion, the FCS- and BSA-free medium with GF-CYK, HA, and RA (M1) showed a comparable development rate to the control medium at 7 and 8 dpf. These growth factors and cytokines in association with hyaluronan and recombinant albumin have a synergistic action by promoting an increase in the blastocyst diameter at 7 dpf. This is fully synthetic method of embryo culture; it presents a valuable tool to reduce the risks of disease transmission via embryo transfer.


2008 ◽  
Vol 15 (11) ◽  
pp. 1666-1673 ◽  
Author(s):  
M. L. V. Watkins ◽  
P. L. Semple ◽  
B. Abel ◽  
W. A. Hanekom ◽  
G. Kaplan ◽  
...  

ABSTRACT Despite routine vaccination with Mycobacterium bovis bacillus Calmette-Guérin (BCG) soon after birth, tuberculosis in babies and adults remains epidemic in South Africa. The immune responses of the naïve newborn child and how they are affected by vaccination with BCG are as yet not fully understood. Immunity during pregnancy and in healthy human newborns may be skewed toward type 2 cytokine production; however, it is type 1 cytokines that are required for protection against M. tuberculosis infection. To better understand neonatal cytokine responses prior to and following exposure to mycobacteria, we have collected cord blood and peripheral blood samples and evaluated the cytokine response following ex vivo incubation with BCG. Gamma interferon (IFN-γ), interleukin 10 (IL-10), IL-12, and low levels of IL-13 and IL-5 but no IL-4 were secreted into the culture supernatant of cord blood mononuclear cells. Intracellular staining showed that IL-10 and IL-12 were produced by monocytes and that IFN-γ was produced by natural killer (NK) cells but not by CD4+ or CD8+ T cells. In contrast, in the peripheral blood samples collected from babies 13 weeks post-BCG vaccination, IFN-γ was detected within CD4+ and CD8+ cells. Taken together, the data suggest a central role for Th1 cytokines in naïve as well as BCG-vaccinated neonates in the protective immune response to tuberculosis. NK cell-derived IFN-γ produced in naïve neonates likely plays a key protective role via monocyte activation and the priming of a subsequent adaptive Th1 response.


2011 ◽  
Vol 52 (9) ◽  
pp. 6138 ◽  
Author(s):  
Leonard Pek-Kiang Ang ◽  
Tien Phuc Do ◽  
Zaw M. Thein ◽  
Hasan Mahmud Reza ◽  
Xiao-Wei Tan ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2844-2844
Author(s):  
Ian K. McNiece ◽  
Jenny Harrington ◽  
Joshua Kellner ◽  
Jennifer Turney ◽  
Elizabeth J. Shpall

Abstract Ex vivo expansion of cord blood products (CB) has been proposed as an approach to increase the number of cells available from a single CB unit. We and others have reported the requirement of CD34 selection for optimal expansion of CB products, however, the selection of frozen CB products results in significant losses of CD34+ cells with a median recovery of 43% (range 6 to 203%, N=40) and low purities resulting in decreased expansion. Therefore we explored approaches to expand CB without prior selection and have described the use of co-culture of CB mononuclear cells (MNC) on mesenchymal stem cells (MSC). In the present study we have evaluated the expansion of clinical CB products (provided by Duke University CB Bank CB). MNC were obtained after ficol separation of RBCs and 10% of the CB product was cultured on preformed layers of MSC in T150 flasks containing 50ml of defined media (Sigma Aldrich) plus 100 ng/ml each of rhSCF, rhG-CSF and rhTpo. After 6 days of culture, the non adherent cells were transferred to a Teflon bag and a further 50 ml of media and GFs added to the flask. Again at day 10, non adherent cells were transferred to the Teflon bag and media and growth factors replaced. At day 12 to 13 of incubation the cells were harvested, washed and total nucleated cell (TNC) counts and progenitor assays performed. In three separate experiments we have achieved greater than 20 fold expansion of TNC with a median of 22, and a median expansion of GM-CFC of 37 fold. Morphologic analysis demonstrated the expanded cells contained high levels of mature neutrophils and neutrophil precursors. In vivo studies in NOD/SCID mice also demonstrated that the expanded cells maintained in vivo engraftment potential. Clinical studies are being designed to evaluate the in vivo potential of CB MNC products expanded on MSC.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3041-3041
Author(s):  
L. R. Fanning ◽  
M. R. Finney ◽  
D. G. Winter ◽  
S. Kadereit ◽  
J. Banks ◽  
...  

Abstract Human CD133+ cells constitute a phenotypically and functionally distinct population of endothelial stem and precursor cells that may play a role in postnatal angiogenesis. CD133+ homing to stimuli, IL-8 production and immunogenic properties are anticipated important characteristics to consider in potential use of allogeneic UCB CD133+ as a therapeutic in arterial ischemia. CD133+ cells were isolated from UCB mononuclear cells (MNC) by magnetic bead selection (AutoMACS, Miltenyi Biotech) according to manufacturer’s protocol and analyzed by flow cytometry. Yields per cord blood unit averaged 1.05 x 106 cells (+/− 0.7 SEM n=30). Surface phenotyping of UCB CD133+ showed co-expression of VEGFR2 (3.5%), CD105 (22.7%) and CXCR4 (8.7%) ligand for SDF-1. Both HLA-DR (57.6%) and HLA-ABC (66.5%) were expressed on CD133+ cells suggesting that CD133+ cells may be capable of presenting immunostimulatory antigens and eliciting an allogeneic reaction. To test this, we performed 96 hour mixed lymphocyte reactions (MLR) using healthy adult peripheral blood MNC as responders stimulated by irradiated (30Gy) CD133+ from UCB (ratio 3:1). Proliferation was measured by 3H-thymidine incorporation. CD133+ and MNC from UCB induced proliferation from allogeneic healthy adult MNC in vitro (46939 +/− 2764 and 49548 +/− 2018 cpm respectively, n=2). MLR studies with CSFE-stained responder cells revealed equivalent rates of lymphocyte cell division comparing selected UCB CD133+ and MNC cells used as stimulators. Comparison studies of responding lymphocyte cytokine production including pro-inflammatory protein assays are ongoing. Initial angiogenic protein assays of CD133+ cells demonstrated elevated levels of IL-8 production as compared to MNC (103+/−380 pg/mL greater in CD133+ than MNC from the same UCB unit) when cultured for 24h in basal media. Transwell migration assays of CD133+ cells to SDF-1 (100ng/mL) demonstrated a 1.8 ± 0.7 fold increase in homing compared to a negative control, coinciding with the CXCR4 expression observed on these cells. In summary, UCB derived CD133+ cells demonstrate homing capability as well as potential for cellular recruitment (IL-8 production) for angiogenesis and cellular therapeutics. CD133+ cells selected from UCB maintain immunostimulatory capacity and initiate proliferation of adult MNC. Further studies of UCB derived CD133+ pro-inflammatory potential; cell recruitment and homing to ischemic signals are warranted.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2203-2203
Author(s):  
Sandeep Chunduri ◽  
Dolores Mahmud ◽  
Javaneh Abbasian ◽  
Damiano Rondelli

Abstract Transplantation of HLA-mismatched cord blood (CB) nucleated cells has limited risk of severe acute graft-versus-host disease and graft rejection. This may depend on naïve T cells not yet exposed to many antigens and on immature antigen-presenting cells (APC) not delivering appropriate signals to allogeneic T cells. In order to test the APC activity of human circulating CB cells in-vitro, we initially used irradiated CB mononuclear cells (MNC) or immunomagnetically selected CD34+ cells, CD133+ cells, or CD14+ monocytes to stimulate the proliferative response of incompatible blood T cells in mixed leukocyte culture (MLC). CB MNC failed to induce allogeneic T cell proliferation, while CD34+ and CD133+ progenitors or CD14+ monocytes induced potent T cell alloresponses. Nevertheless, since allogeneic T cell response was not restored after depletion of CD3+ cells in the CB, nor the add-back of irradiated CB MNC to CD34+ or CD14+ stimulators inhibited allo-T cells, a direct suppressive effect of CB MNC was excluded. Allogeneic peripheral blood cytotoxic T-lymphocyte (CTL) responses were not induced after 7 days of stimulation with irradiated CB MNC, although after 4 weekly rechallenges with CB MNC, on average a 23% lysis of antigen-specific CB PHA-blasts was observed at the highest effector:target ratio (50:1). To test the tolerogenic potential of CB MNC, T cells initially exposed to CB MNC were rechallenged in secondary MLC with CB MNC, or CD34+ cells, or monocyte-derived dendritic cells (Mo-DC) generated in liquid culture with GM-CSF and IL-4. Allogeneic T cells were still unresponsive upon rechallenge with CB MNC, but proliferated upon 3 days of restimulation with CD34+ cells or Mo-DC from the same CB. Surprisingly, the supernatant of these latter MLCs did inhibit completely a 3rd party MLC. Instead, the supernatant of blood T cells that had been activated by CB CD34+ cells or Mo-DC both in primary and secondary MLC did not. These results show an impaired allo-APC activity of CB MNC but not CB CD34+ cells, and suggest that T cells releasing immunosuppressive cytokines may be activated by CB MNC and then expanded by a second more potent stimulation with professional APC. This hypothesis could explain the sustained engraftment of HLA-mismatched CB stem cell transplants in humans. Based on these results, the in-vivo or ex-vivo downregulation of T cell alloreactivity induced by CB MNC will be tested in experimental models of stem cell, as well as solid organ transplantation.


Sign in / Sign up

Export Citation Format

Share Document