Expansion of Hematopoietic Stem Cells (HSC) from Cord-Blood (CB) Derived Mononuclear Cells (MNC) in Cytokine-Free Environment Using Mesenchymal Cells Spatial Co-Culture System.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2565-2565
Author(s):  
Shai Meretzki ◽  
Ora Burger ◽  
Osnat Merom-Jacov ◽  
Avinoam Kaduri ◽  
Jacob M. Rowe ◽  
...  

Abstract In situ, HSCs are intimately associated with discrete spatially organized niches within the bone marrow, which are part of the “hematopoietic inductive microenvironment” (HIM). The HIM provides a range of molecular signals that collectively control HSC differentiation and self-renewal. This process is mediated via cell-cell and cell-ECM molecular contacts or through specific factors synthesized and secreted by mesenchymal stromal cells (MSC). Current strategies aimed at ex vivo expansion of transplantable HSC have so far been met with limited success. Most attempts to expand HSC ex-vivo are based on using monolayers of MSC as a supportive tier or growth media supplemented with blend of cytokines. failure to support long-term maintenance and expansion of human HSC on MSC monolayers could be associated with inadequate physical architecture of the culture systems which does not reflect the natural 3-D growth conditions present within the BM-HIM. The other method based on supporting the Hematopoietic cells proliferation by a blend of cytokines, is proven routine but post transplantation marrow replenishment is currently unsatisfactory and chromosomal epigenetic modifications may be introduced into progeny cells. Using the PluriX™ bioreactor, we had previously demonstrated that spatial co-cultures of MSC and HSCs provide an efficient system for the expansion of HSCs from CB CD34+ selected cells in cytokine-free environment. Over the past years, attempts to expand HSCs mostly employed CD34+ selected cells. However, these cells may not represent the earliest HSCs and the immunomagnetic selection protocols are costly, time consuming and associated with great loss of source cells. A theoretical approach to overcome both hurdles is by using non-selected MNC as the founding population of HSC. Following our ability to expand HSCs from CD34+ selected cells, we now demonstrate the capacity of the 3-D HSC MSCs co-culture system within the PluriX™ bioreactor system to expand HSCs using MNC as HSCs source. Primary Human marrow-derived MSCs were grown on 3-D carriers within the PluriX™bioreactor system. When the MSCs cultures reached high density (3*106–8*106 cells/ml) CD34+ selected cells or MNC were plated onto them. Within 7–14 days, the population of CD34+ and CD34+CD38− cell were expanded irrespective of the HSCs founding source. However, during this period of time the absolute expansion magnitude was greater when MNC rather than CD34+ selected cells were used to drive the process. The enrichment rate of CD34+cell from MNC and CD34+ selected cells was 20–100 vs. 5–20 fold, respectively. Under same conditions, the enrichment of the earlier CD34+CD38− cells was 40–50 vs. up to 10 fold for the same cell populations. In-conclusion, the spatial co-cultures of MSC and HSC within the PluriX™ bioreactor have been shown to form a potent HSCs expansion system in non-supplemented cytokine environment. The capacity of this practice is improved when MNCs, instead of CD34+ selected cells, serve as the originating population for HSCs. It is conceivable that the efficiency of this system is based upon selective adherence between HSCs and the hematopoietic niches within the MSCs 3-D cultures. As such, upscale of the current system could become attractive method for HSCs expansion.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2865-2865
Author(s):  
Osamu Imataki ◽  
Yuji Heike ◽  
Akira Iiduka ◽  
Masasi Kuwatani ◽  
Yoshinori Ikarashi ◽  
...  

Abstract Background: Natural killer T (NKT) cells are one of the primary effectors of the innate immune systems, and also have an important role to initiate and regulate adaptive immune responses. Previously, we reported that Vα24+ NKT cells proliferated more efficiently from granulocyte-colony stimulating factor (G-CSF)-mobilized peripheral blood mononuclear cells (PBMC) compared with steady-state blood cells. However, optimal culture conditions and characterization of ex vivo expanded NKT cells have not been fully evaluated. Object: We examined several different conditions including cell ratios, mediums, growth factors and incubation schedules to seek an optimal culture system for expansion of NKT cells. Methods: PBMC were collected from donors for hematopoietic stem cell transplantation before and after G-CSF administration, and were cultured in AIM-V medium supplemented with 10% auto-plasma, 100 ng/mL α-galactosylceramide (α-GalCer) and 100 U/mL recombinant human (rh) IL-2. IL-2 alone was repeatedly charged every 3 days to maintain its biological activity. After 12 days culture, we compared the expansion efficacy of Vα24+CD3+ NKT cells derived from PBMC with or without G-CSF. For depletion analysis, we used a magnetic cell sorting (MACS) system with labeling magnetic beads-conjugated monoclonal antibody against CD14, 56, 34, and TCR Vα24 chain. Results: The expansion fold of Vα24+CD3+ NKT cells were significantly higher with G-CSF (669 vs 182 fold, n=20). Among cell populations we tested, the proportion of CD14+CD16+ cells before cultures were associated with the efficacy of Vα24+CD3+ NKT cells expansion, and the proportion of CD34+, Vα24+, CD56+ and CD56+CD161+ cells were not. The magnitude of expansion of Vα24+CD3+ NKT cells was correlated with the percentage of CD14+ cells at the initiation of cultures. Proliferation of Vα24+CD3+ NKT cells was abrogated by the depletion of Vα24+cells, but notCD34+ cells. Depletion of CD56+ T cells induced higher expansion ratio of Vα24+CD3+ NKT, which was abolished when CD56+ and CD56− cells were cultured separately using a 3 μm pored-membrane filter. Furthermore, co-culture of enriched Vα24+ cells and purified CD56+ cells inhibited the proliferation of Vα24+CD3+ NKT cells. It was hypothesized that the repeated IL-2 supplementation resulted in enhancement of CD56+ cells (NK cells) to suppress the proliferation of Vα24+CD3+ NKT cells. We tested different administration schedule of IL-2 as follow: on day 0 only, day 0 & 3, day 0, 3 & 6, day 0, 3, 6 & 9 (that is every 3 days), and we found that Vα24+CD3+ NKT cells expanded most effectively when IL-2 was supplemented on day 0 only. In order to modify the number of CD14+ cells in culture system, we added back CD14+ cells to CD14− cells culture on day 0, 3, 6, 9 or every 3 days, but this did not result in significant enhancement of proliferation of Vα24+CD3+ NKT cells. Conclusions: For efficient ex vivo culture of Vα24+ NKT cells, the presence of Vα24+ NKT cells and CD14+ cells are critical. The NK cells may interfere the interaction between antigen presenting cells (APC) and NKT cells by hindering a function of antigen presentation or yielding direct cytotoxicity against APC. These findings are helpful to develop an efficient expansion system of NKT cells in feature adaptive immunotherapy.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Rupak Dua ◽  
Hugh Jones ◽  
Philip C. Noble

AbstractRecent advances in materials and manufacturing processes have allowed the fabrication of intricate implant surfaces to facilitate bony attachment. However, refinement and evaluation of these new design strategies are hindered by the cost and complications of animal studies, particularly during early iterations in the development process. To address this problem, we have previously constructed and validated an ex-vivo bone bioreactor culture system that can maintain the viability of bone samples for an extended period ex-vivo. In this study, we investigated the mineralization of a titanium wire mesh scaffold under both static and dynamic culturing using our ex vivo bioreactor system. Thirty-six cancellous bone cores were harvested from bovine metatarsals at the time of slaughter and divided into five groups under the following conditions: Group 1) Isolated bone cores placed in static culture, Group 2) Unloaded bone cores placed in static culture in contact with a fiber-mesh metallic scaffold, Group 3) Bone cores placed in contact with a fiber-mesh metallic scaffold under the constant pressure of 150 kPa, Group 4) Bone core placed in contact with a fiber-mesh metallic scaffold and exposed to cyclic loading with continuous perfusion flow of media within the ex-vivo culture system and Group 5) Bone core evaluated on Day 0 to serve as a positive control for comparison with all other groups at weeks 4 and 7. Bone samples within Groups 1–4 were incubated for 4 and 7 weeks and then evaluated using histological examination (H&E) and the Live-Dead assay (Life Technologies). Matrix deposits on the metallic scaffolds were examined with scanning electron microscopy (SEM), while the chemical composition of the matrix was measured using energy-dispersive x-ray spectroscopy (EDX). We found that the viability of bone cores was maintained after seven weeks of loading in our ex vivo system. In addition, SEM images revealed crystallite-like structures on the dynamically loaded metal coupons (Group 4), corresponding to the initial stages of mineralization. EDX results further confirmed the presence of carbon at the interface and calcium phosphates in the matrix. We conclude that a bone bioreactor can be used as an alternate tool for in-vivo bone ingrowth studies of new implant surfaces or coatings.


Blood ◽  
1999 ◽  
Vol 94 (5) ◽  
pp. 1623-1636 ◽  
Author(s):  
Chu-Chih Shih ◽  
Mickey C.-T. Hu ◽  
Jun Hu ◽  
Jeffrey Medeiros ◽  
Stephen J. Forman

Abstract We have developed a stromal-based in vitro culture system that facilitates ex vivo expansion of transplantable CD34+thy-1+ cells using long-term hematopoietic reconstitution in severe combined immunodeficient-human (SCID-hu) mice as an in vivo assay for transplantable human hematopoietic stem cells (HSCs). The addition of leukemia inhibitory factor (LIF) to purified CD34+ thy-1+ cells on AC6.21 stroma, a murine bone marrow–derived stromal cell line, caused expansion of cells with CD34+ thy-1+ phenotype. Addition of other cytokines, including interleukin-3 (IL-3), IL-6, granulocyte-macrophage colony-stimulating factor, and stem cell factor, to LIF in the cultures caused a 150-fold expansion of cells retaining the CD34+ thy-1+ phenotype. The ex vivo–expanded CD34+ thy-1+ cells gave rise to multilineage differentiation, including myeloid, T, and B cells, when transplanted into SCID-hu mice. Both murine LIF (cannot bind to human LIF receptor) and human LIF caused expansion of human CD34+ thy-1+ cells in vitro, suggesting action through the murine stroma. Furthermore, another human HSC candidate, CD34+ CD38− cells, shows a similar pattern of proliferative response. This suggests thatex vivo expansion of transplantable human stem cells under this in vitro culture system is a general phenomenon and not just specific for CD34+ thy-1+ cells.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4147-4147
Author(s):  
Sonja Loges ◽  
Martin Butzal ◽  
Uta Fischer ◽  
Ursula M. Gehling ◽  
Dieter K. Hossfeld ◽  
...  

Abstract The rare CD133+ stem cell population contains both hematopoietic and endothelial progenitors. Successful ex-vivo expansion of this multipotent population would therefore be of great benefit in many clinical settings including stem cell transplantation and gene therapy. We developed a cell culture system containing the recombinant human cytokines vascular endothelial growth factor (VEGF), FLT3 ligand (FLT3L) and stem cell growth factor (SCGF) for ex-vivo expansion of purified human CD133+ stem cells obtained from leukapheresis products from patients pre-treated with G-CSF. FACS analysis, colony assays and NOD-SCID transplantation studies were performed to monitor stem cell and endothelial phenotype in-vitro and in-vivo. Cultivation with VEGF, FLT3L and SCGF induced a mean 2200-fold increase of total cell counts in 5 weeks. FACS analysis revealed persistence of 6–15% CD133+ stem cells indicating proliferation and survival of primitive hematopoietic stem cells. 5–6% of the proliferating cells expressed the endothelial markers CD144 (VE-Cadherin) and von-Willebrand factor (vWF). Ex-vivo expanded stem cells could be differentiated into adherent endothelial cells after withdrawal of SCGF and FLT3L allowing generation of large numbers of endothelial cells. Colony-assays showed an increase of hematopoietic and endothelial colonies after 5 weeks of ex-vivo expansion indicating simultaneous proliferation of hematopoietic and endothelial precursors under the established culture conditions (CFU-E 60-fold, CFU-GEMM 48-fold, CFU-GM 59-fold, CFU-G 99-fold, CFU-M 1356-fold and CFU-EC 1843-fold). To assess in-vivo functionality, hematopoietic stem cells expanded ex-vivo for 7, 14, 21 and 32 days were transplanted into sublethally irradiated NOD-SCID mice. For each expansion period, the mean percentage of anti-human CD45 positive bone marrow cells 3 months post-transplantation was 11, 3, 3 and 1%, respectively. Human CD45+ cells for each set of experiments contained a mean of 15, 26, 8 and 32% T-cells (CD3+), 9, 0, 7 and 21% B-cells (CD19+), 24, 2, 2 and 11% monocytes (CD14+), 21, 3, 1 and 12% granulocytes (CD33+) and 19, 37, 44 and 24% stem cells (CD34+) (d7 (n=5), d14 (n=4), d21 (n=7) and d32 (n=6) respectively). Our experiments showed multilineage engraftment of human stem cells expanded for more than 4 weeks ex-vivo. Therefore our culture system provides a tool to generate large numbers of human stem and endothelial cells for clinical purposes.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1068-1068
Author(s):  
Naoko Takebe ◽  
Thomas MacVittie ◽  
Xiangfei Cheng ◽  
Ann M. Farese ◽  
Emily Welty ◽  
...  

Abstract Down-modulation of surface CXCR4, a G-protein-coupled receptor, in hematopoietic stem cells (HSCs) undergoing ex vivo expansion culturing is considered to be one of the major causes of marrow reconstitution failure, possibly due to an HSC homing defect. Recently, it has been reported that severe combined immunodeficiency (SCID)-repopulating cells (SRC) were expanded from the CD34-enriched human adult bone marrow (ABM) or cord blood (CB) hematopoietic stem cells (HSC) using a human brain endothelial cell (HUBEC) co-culture system. We found that primitive cord blood cells expressing surface CXCR4 (82+5%) lost this capability significantly during 7 days of ex vivo expansion in the HUBEC co-culture containing the cytokines stem cell factor (SCF), flt-3, interleukin (IL)-6, IL-3, and granulocyte macrophage colony stimulating factor (GM-CSF). Expression levels of other surface proteins relevant to HSC homing, such as CD49d, CD95, CD26, or CD11a, were not down-modulated. We hypothesized that CXCR4 down-regulation was caused by a receptor internalization and tested several methods to reverse CXCR4 internalization back to the surface, such as elimination of GM-CSF in the culture media, performing a non-contact culture using the transwell, or adding either 0.3Mor 0.4M sucrose, or 25μg/ml chlorpromazine (CPZ), 24 hours prior to the analysis. CPZ and sucrose are known inhibitors of the cytokine-induced endocytosis of CXCR4 in neutrophils (Bruhl H. et al. Eur J Immunol 2003). Interestingly, 0.4M sucrose showed approximately a 2-fold increase of surface CXCR4 expression on CB CD34+ cells by flow cytometry analysis. CPZ and 0.3M sucrose showed a moderate increase expression of CXCR4. Using a transwell HUBEC co-culture system, CXCR4 surface expression on CD34+ cells was down-regulated during the ex vivo culture. In vitro HSC migration test showed 3.1-fold increase in migration compared to the control after incubation of HSC with 0.1M sucrose for 16 hours prior to the in vitro migration study. Eliminating GM-CSF from the cytokine cocktail or adding MG132 increased migration 1.36- and 1.2-fold compared to the control. We are currently performing an in vivo homing assay using nonobese diabetic (NOD)-SCID mice. In conclusion, the HUBEC ex vivo culture system down-regulates surface CXCR4 in human cord blood HSC. The mechanism of CXCR4 surface down regulation may be receptor internalization by cytokines. Sucrose may be useful in attenuation of CXCR4 surface expression in CD34+ HSC by inhibition of receptor internalization via clathrin-coated pits.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 341-341
Author(s):  
Teresa Mortera Blanco ◽  
Athanasios Mantalaris ◽  
Joseph Santiapillai ◽  
Alexander Bismarck ◽  
Nicki Panoskaltsis

Abstract Abstract 341 Ex vivo expansion of cord blood mononuclear cells (CBMNCs) could provide a safe, flexible and ample supply of blood components for cellular therapies. Traditionally, hematopoietic cell expansion has been performed in 2D tissue culture flask or well-plate static cultures using abnormally high concentrations of cytokines which is expensive, reduces the self-renewal capacity, and skews normal differentiation. We have previously developed a 3D bone marrow biomimicry through the use of a synthetic scaffold made of polyurethane (PU) coated with collagen type I which could expand CBMNCs in a cytokine-free environment for at least 28 days ex vivo, with or without the addition of serum to the media. We hypothesised that the addition of near physiological concentrations (0.2U/mL and 1.845U/mL) of exogenous erythropoietin (EPO) to these established 3D CBMNC ex vivo cultures at day 14 in a serum-free and cytokine-free environment would be sufficient to enhance erythropoiesis. CBMNCs were separated by Ficoll-Paque density gradient and seeded onto collagen-coated PU 3D scaffolds at a cell density of 2.5×106cells per scaffold (5×5×5mm3). Cultures were established in serum-free conditions and only EPO was added at days 14–28, with full-medium exchange every 2 days. Culture output was evaluated at days 14, 21 and 28 both by physically extracting cells from the scaffolds and by in situ analysis. Over 28 days, most stages of maturation, from erythroid progenitors to enucleated erythrocytes were observed by light microscopy of cytospins and by immunophenotypic analysis of extracted cells (CD45−/CD71+/CD235+), with more maturation occurring by day 28 of culture, after the addition of EPO. Although both concentrations of EPO produced comparable erythroid differentiation of cells, even by CFU assay, the viability (75% vs. 61%, p<0.05) and proliferative capacity at day 28 of culture was enhanced in the higher concentration of EPO compared with that in the lower concentration (p<0.05). In contrast, standard 2D control cultures (without serum or cytokines) collapsed within 5 days. In situ, scanning electron microscopy showed maturation of erythrocytes within central sections of the scaffolds to enucleation by day 28 and multiphoton microscopy confirmed the presence of structures resembling erythroid islands as early as day 14 of culture, prior to the addition of EPO. In conclusion, 3D PU-collagen scaffolds may provide a good model to study erythropoiesis ex vivo, using physiological concentrations of EPO, and has the potential to expand red cells in response to higher levels of exogenous EPO in a culture system that would be suitable for clinical applications. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1999 ◽  
Vol 94 (5) ◽  
pp. 1623-1636 ◽  
Author(s):  
Chu-Chih Shih ◽  
Mickey C.-T. Hu ◽  
Jun Hu ◽  
Jeffrey Medeiros ◽  
Stephen J. Forman

We have developed a stromal-based in vitro culture system that facilitates ex vivo expansion of transplantable CD34+thy-1+ cells using long-term hematopoietic reconstitution in severe combined immunodeficient-human (SCID-hu) mice as an in vivo assay for transplantable human hematopoietic stem cells (HSCs). The addition of leukemia inhibitory factor (LIF) to purified CD34+ thy-1+ cells on AC6.21 stroma, a murine bone marrow–derived stromal cell line, caused expansion of cells with CD34+ thy-1+ phenotype. Addition of other cytokines, including interleukin-3 (IL-3), IL-6, granulocyte-macrophage colony-stimulating factor, and stem cell factor, to LIF in the cultures caused a 150-fold expansion of cells retaining the CD34+ thy-1+ phenotype. The ex vivo–expanded CD34+ thy-1+ cells gave rise to multilineage differentiation, including myeloid, T, and B cells, when transplanted into SCID-hu mice. Both murine LIF (cannot bind to human LIF receptor) and human LIF caused expansion of human CD34+ thy-1+ cells in vitro, suggesting action through the murine stroma. Furthermore, another human HSC candidate, CD34+ CD38− cells, shows a similar pattern of proliferative response. This suggests thatex vivo expansion of transplantable human stem cells under this in vitro culture system is a general phenomenon and not just specific for CD34+ thy-1+ cells.


2005 ◽  
Vol 33 (7) ◽  
pp. 828-835 ◽  
Author(s):  
Cláudia Lobato da Silva ◽  
Raquel Gonçalves ◽  
Kirsten B. Crapnell ◽  
Joaquim M.S. Cabral ◽  
Esmail D. Zanjani ◽  
...  

Blood ◽  
2000 ◽  
Vol 95 (6) ◽  
pp. 1957-1966 ◽  
Author(s):  
Chu-Chih Shih ◽  
Mickey C.-T. Hu ◽  
Jun Hu ◽  
Yehua Weng ◽  
Paul J. Yazaki ◽  
...  

Abstract The development of culture systems that facilitate ex vivo maintenance and expansion of transplantable hematopoietic stem cells (HSCs) is vital to stem cell research. Establishment of such culture systems will have significant impact on ex vivo manipulation and expansion of transplantable stem cells in clinical applications such as gene therapy, tumor cell purging, and stem cell transplantation. We have recently developed a stromal-based culture system that facilitates ex vivo expansion of transplantable human HSCs. In this stromal-based culture system, 2 major contributors to the ex vivo stem cell expansion are the addition of leukemia inhibitory factor (LIF) and the AC6.21 stromal cells. Because the action of LIF is indirect and mediated by stromal cells, we hypothesized that LIF binds to the LIF receptor on AC6.21 stromal cells, leading to up-regulated production of stem cell expansion promoting factor (SCEPF) and/or down-regulated production of stem cell expansion inhibitory factor (SCEIF). Here we demonstrate a secreted SCEPF activity in the conditioned media of LIF-treated AC6.21 stromal cell cultures (SCM-LIF). The magnitude of ex vivo stem cell expansion depends on the concentration of the secreted SCEPF activity in the SCM-LIF. Furthermore, we have ruled out the contribution of 6 known early-acting cytokines, including interleukin-3, interleukin-6, granulocyte macrophage colony-stimulating factor, stem cell factor, flt3 ligand, and thrombopoietin, to this SCEPF activity. Although further studies are required to characterize this secreted SCEPF activity and to determine whether this secreted SCEPF activity is mediated by a single factor or by multiple growth factors, our results demonstrate that stromal cells are not required for this secreted SCEPF activity to facilitate ex vivo stem cell expansion.


Author(s):  
André Branco ◽  
Sara Bucar ◽  
Jorge Moura-Sampaio ◽  
Carla Lilaia ◽  
Joaquim M. S. Cabral ◽  
...  

Umbilical cord blood (UCB) has been established as an alternative source for hematopoietic stem/progenitor cells (HSPC) for cell and gene therapies. Limited cell yields of UCB units have been tackled with the development of cytokine-based ex vivo expansion platforms. To improve the effectiveness of these platforms, namely targeting clinical approval, in this study, we optimized the cytokine cocktails in two clinically relevant expansion platforms for HSPC, a liquid suspension culture system (CS_HSPC) and a co-culture system with bone marrow derived mesenchymal stromal cells (BM MSC) (CS_HSPC/MSC). Using a methodology based on experimental design, three different cytokines [stem cell factor (SCF), fms-like tyrosine kinase 3 ligand (Flt-3L), and thrombopoietin (TPO)] were studied in both systems during a 7-day culture under serum-free conditions. Proliferation and colony-forming unit assays, as well as immunophenotypic analysis were performed. Five experimental outputs [fold increase (FI) of total nucleated cells (FI TNC), FI of CD34+ cells, FI of erythroid burst-forming unit (BFU-E), FI of colony-forming unit granulocyte-monocyte (CFU-GM), and FI of multilineage colony-forming unit (CFU-Mix)] were followed as target outputs of the optimization model. The novel optimized cocktails determined herein comprised concentrations of 64, 61, and 80 ng/mL (CS_HSPC) and 90, 82, and 77 ng/mL (CS_HSPC/MSC) for SCF, Flt-3L, and TPO, respectively. After cytokine optimization, CS_HSPC and CS_HSPC/MSC were directly compared as platforms. CS_HSPC/MSC outperformed the feeder-free system in 6 of 8 tested experimental measures, displaying superior capability toward increasing the number of hematopoietic cells while maintaining the expression of HSPC markers (i.e., CD34+ and CD34+CD90+) and multilineage differentiation potential. A tailored approach toward optimization has made it possible to individually maximize cytokine contribution in both studied platforms. Consequently, cocktail optimization has successfully led to an increase in the expansion platform performance, while allowing a rational side-by-side comparison among different platforms and enhancing our knowledge on the impact of cytokine supplementation on the HSPC expansion process.


Sign in / Sign up

Export Citation Format

Share Document