scholarly journals Stem cell integrins: Implications for ex-vivo culture and cellular therapies

2011 ◽  
Vol 6 (1) ◽  
pp. 1-12 ◽  
Author(s):  
Andrew B.J. Prowse ◽  
Fenny Chong ◽  
Peter P. Gray ◽  
Trent P. Munro
Blood ◽  
1996 ◽  
Vol 87 (11) ◽  
pp. 4589-4595 ◽  
Author(s):  
TL Holyoake ◽  
MG Freshney ◽  
L McNair ◽  
AN Parker ◽  
PJ McKay ◽  
...  

The characterization of many cytokines involved in the control of hematopoiesis has led to intense investigation into their potential use in ex vivo culture to expand progenitor numbers. We have established the optimum ex vivo culture conditions that allow substantial amplification of transient engrafting murine stem cells and which, simultaneously, augment the ability to sustain serial bone marrow transplantation (BMT). Short-term incubation of unfractionated BM cells in liquid culture with stem cell factor (SCF) and interleukin-11 (IL- 11) produced a 50-fold amplification of clonogenic multipotential progenitors (CFU-A). Following such ex vivo expansion, substantially fewer cells were required to rescue lethally irradiated mice. When transplanted in cell doses above threshold for engraftment, BM cells expanded ex vivo resulted in significantly more rapid hematopoietic recovery. In a serial transplantation model, unmanipulated BM was only able to consistently sustain secondary BMT recipients, but BM expanded ex vivo has sustained quaternary BMT recipients that remain alive and well more than 140 days after 4th degree BMT. These results show augmentation of both short-term recovery posttransplant and the ability to serially transplant marrow by preincubation in culture with SCF and IL-11.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5545-5545
Author(s):  
Brandon K. Wyss ◽  
Justin L. Meyers ◽  
Anthony L. Sinn ◽  
Shanbao Cai ◽  
Karen E. Pollok ◽  
...  

Abstract We previously demonstrated that engraftment of murine whole bone marrow (WBM) transduced with an oncoretroviral vector using an optimized 5-fluorouracil (5-FU)-based transduction protocol is reduced ~3-fold compared to fresh WBM upon transplantation into sublethally irradiated hosts, although competitive repopulating ability in ablated hosts is not decreased (Goebel et al., Exp. Hematol. 30:1324, 2002). We therefore sought to determine whether marrow cells transduced using a clinically relevant, non-5-FU-containing protocol would engraft more efficiently. Li et al. (Exp. Hematol. 31:1206, 2003) showed that lineage-depleted (lin−) marrow cells from donor mice not treated with 5-FU were effectively transduced, and repopulated myeloablated hosts. We hypothesized that ex vivo culture for gene transfer in the absence of 5-FU would lead to improved donor marrow engraftment in submyeloablated hosts. Lin− cells, isolated from B6.SJL (Boy J; CD45.1+) WBM using a Miltenyi kit and VarioMACS apparatus, were prestimulated in StemSpan serum-free medium with SCF and IL-6 for 48 hours, followed by overnight transduction on RetroNectin-coated plates preloaded with ecotropic SF1-EGFP retroviral supernatant. Cell recovery from the MACS column (1.9 ± 1.4%), bulk transduction efficiency (77.3 ± 12%), and lin− cell purity (58 ± 17%; all from 7–13 experiments) was similar to that previously described. Transplantation of 106 lin− transduced cells into 300 cGy-conditioned congenic C57Bl6/J (B6; CD45.2+) hosts produced only 1.4 ± 0.5% donor chimerism 4–6 months post-transplant, significantly lower than that observed using 106 fresh lin− cells (29 + 18.8%; N = 8–9 hosts each from 2–3 experiments). The percentage of EGFP+ cells in the donor population, nevertheless, was 55.6 ± 18%, indicating that stem cells were marked but engrafted poorly. The repopulating ability of transduced lin− marrow was reduced ~10-fold compared to fresh lin− cells as determined in competitive repopulation assays in ablated hosts. Together, these data suggest that lin cells cultured ex vivo for gene transfer acquired an engraftment defect despite the absence of 5-FU. Increasing the conditioning radiation dose to 550 cGy, a dose used in prior canine and non-human primate gene transfer studies, markedly improved donor chimerism following transplantation of 106 fresh lin− cells (90 ± 1.3% at 4 months, N = 5) or 106 transduced lin− cells (38.5 ± 14% at 2 months, N = 10), suggesting that greater reduction in host stem cell function may be needed for engraftment of cells cultured ex vivo for gene transfer. Ongoing studies to investigate the mechanism responsible for this engraftment defect indicate that expression of adhesion molecules important for homing and engraftment (CD29, 44, 49d, 49e, 62L), CXCR4 expression, and the percentage of cells actively cycling are not significantly altered by the transduction process, although functional studies are underway. The percentage of Sca-1+lin−c-kit+ (SLK) cells in the transduced cell pool is similar to that of freshly isolated lin− cells; thus, transplantation of lin− cells cultured ex vivo for gene transfer results in significantly lower donor chimerism than fresh lin− cells despite the grafts containing similar numbers of SLK cells. Secondary transplants and limiting dilution studies to determine stem cell self-renewal and engraftment capacity before and after ex vivo culture for gene transfer are in progress.


Blood ◽  
1998 ◽  
Vol 91 (1) ◽  
pp. 111-117 ◽  
Author(s):  
Dimitri A. Breems ◽  
Ellen A.W. Blokland ◽  
Karen E. Siebel ◽  
Angelique E.M. Mayen ◽  
Lilian J.A. Engels ◽  
...  

Stroma-supported long-term cultures (LTC) allow estimation of stem cell quality by simultaneous enumeration of hematopoietic stem cell (HSC) frequencies in a graft using the cobblestone area forming cell (CAFC) assay, and the ability of the graft to generate progenitors in flask LTC (LTC-CFC). We have recently observed that the number and quality of mobilized peripheral blood stem cells (PBSC) was low in patients having received multiple rounds of chemotherapy. Moreover, grafts with low numbers of HSC and poor HSC quality had a high probability to cause graft failure upon their autologous infusion. Because ex vivo culture of stem cells has been suggested to present an attractive tool to improve hematological recovery or reduce graft size, we have studied the possibility that such propagation may affect stem cell quality. In order to do so, we have assessed the recovery of different stem cell subsets in CD34+ PBSC after a 7-day serum-free liquid culture using CAFC and LTC-CFC assays. A numerical expansion of stem cell subsets was observed in the presence of interleukin-3 (IL-3), stem cell factor, and IL-6, while stroma-contact, stromal soluble factors, or combined addition of FLT3-ligand and thrombopoietin improved this parameter. In contrast, ex vivo culture severely reduced the ability of the graft to produce progenitors in LTC while stromal soluble factors partly abrogated this quality loss. The best conservation of graft quality was observed when the PBSC were cultured in stroma-contact. These data suggest that ex vivo propagation of PBSC may allow numerical expansion of various stem cell subsets, however, at the expense of their quality. In addition, cytokine-driven PBSC cultures require stroma for optimal maintenance of graft quality.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1677-1677
Author(s):  
Zejin Sun ◽  
Yanzhu Yang ◽  
Yan Li ◽  
Daisy Zeng ◽  
Jingling Li ◽  
...  

Abstract Fanconi anemia (FA) is a recessive DNA repair disorder characterized by congenital abnormalities, bone marrow failure, genomic instability, and a predisposition to malignancies. As the majority of FA patients ultimately acquires severe bone marrow failure, transplantation of stem cells from a normal donor is the only curative treatment to replace the malfunctioning hematopoietic system. Stem cell gene transfer technology aimed at re-introducing the missing gene is a potentially promising therapy, however, prolonged ex vivo culture of cells, that was utilized in clinical trials with gammaretroviruses, results in a high incidence of apoptosis and at least in mice predisposes the surviving reinfused cells to hematological malignancy. Consequently, gene delivery systems such as lentiviruses that allow a reduction in ex vivo culture time are highly desirable. Here, we constructed a lentiviral vector expressing the human FANCA cDNA and tested the ability of this construct pseudotyped with either VSVG or a modified prototype foamyvirus (FV) envelope to correct Fanca−/− stem and progenitor cells in vitro and in vivo. In order to minimize genotoxic stress due to extended in vitro manipulations, an overnight transduction protocol was utilized where in the absence of prestimulation, murine Fanca−/− bone marrow cKit+ cells were co-cultured for 16h with FANCA lentivirus on the recombinant fibronectin fragment CH296. Transduction efficiency and transfer of lentivirally expressed FANCA was confirmed functionally in vitro by improved survival of consistently approximately 60% of clonogenic progenitors in serial concentrations of mitomycin C (MMC), irregardless of the envelope that was utilized to package the vector. Transduction of fibroblasts was also associated with complete correction of MMC-induced G2/M arrest and biochemically with the restoration of FancD2 mono-ubiquitination. Finally, to functionally determine whether gene delivery by the recombinant lentivirus during such a short transduction period is sufficient to correct Fanca−/− stem cell repopulation to wild-type levels, competitive repopulation experiments were conducted as previously described. Follow-up of up to 8 months demonstrated that the functional correction were also achieved in the hematopoietic stem cell compartment as evidenced by observations that the repopulating ability of Fanca−/− stem cells transduced with the recombinant lentivirus encoding hFANCA was equivalent to that of wild-type stem cells. Importantly, despite the fact that the gene transfer efficiency into cells surviving the transduction protocol were similar for both pseudotypes, VSVG was associated with a 4-fold higher toxicity to the c-kit+ cells than the FV envelope. Thus, when target cell numbers are limited as stem cells are in FA patients, the foamyviral envelope may facilitate overall greater survival of corrected stem cells. Collectively, these data indicate that the lentiviral construct can efficiently correct FA HSCs and progenitor cells in a short transduction protocol overnight without prestimulation and that the modified foamy envelope may have less cytotoxicity than the commonly used VSVG envelope.


Cytotherapy ◽  
2014 ◽  
Vol 16 (4) ◽  
pp. S60
Author(s):  
C.L. da Silva ◽  
P.Z. Andrade ◽  
A.M. Soure ◽  
F. Dos Santos ◽  
G. Almeida-Porada ◽  
...  

Blood ◽  
1998 ◽  
Vol 91 (1) ◽  
pp. 111-117 ◽  
Author(s):  
Dimitri A. Breems ◽  
Ellen A.W. Blokland ◽  
Karen E. Siebel ◽  
Angelique E.M. Mayen ◽  
Lilian J.A. Engels ◽  
...  

Abstract Stroma-supported long-term cultures (LTC) allow estimation of stem cell quality by simultaneous enumeration of hematopoietic stem cell (HSC) frequencies in a graft using the cobblestone area forming cell (CAFC) assay, and the ability of the graft to generate progenitors in flask LTC (LTC-CFC). We have recently observed that the number and quality of mobilized peripheral blood stem cells (PBSC) was low in patients having received multiple rounds of chemotherapy. Moreover, grafts with low numbers of HSC and poor HSC quality had a high probability to cause graft failure upon their autologous infusion. Because ex vivo culture of stem cells has been suggested to present an attractive tool to improve hematological recovery or reduce graft size, we have studied the possibility that such propagation may affect stem cell quality. In order to do so, we have assessed the recovery of different stem cell subsets in CD34+ PBSC after a 7-day serum-free liquid culture using CAFC and LTC-CFC assays. A numerical expansion of stem cell subsets was observed in the presence of interleukin-3 (IL-3), stem cell factor, and IL-6, while stroma-contact, stromal soluble factors, or combined addition of FLT3-ligand and thrombopoietin improved this parameter. In contrast, ex vivo culture severely reduced the ability of the graft to produce progenitors in LTC while stromal soluble factors partly abrogated this quality loss. The best conservation of graft quality was observed when the PBSC were cultured in stroma-contact. These data suggest that ex vivo propagation of PBSC may allow numerical expansion of various stem cell subsets, however, at the expense of their quality. In addition, cytokine-driven PBSC cultures require stroma for optimal maintenance of graft quality.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2113-2113
Author(s):  
Peter Kurre ◽  
Ponni Anandakumar ◽  
Vladimir A. Lesnikov ◽  
Hans-Peter Kiem

Abstract Most gene transfer models using Moloney murine leukemia virus (MLV) - derived vectors to target hematopoietic repopulating cells require progenitor cell enrichment and extended ex vivo culture for efficient long-term marking. Both may result in qualitative, and/or quantitative, loss of stem cells thereby limiting gene transfer rates in vivo. This can be a critical obstacle in candidate applications with exhausted autologous stem cell pools, such as Fanconi Anemia. Among the advantages of HIV-derived lentivirus vectors is their ability to transduce non dividing cells, permitting shortened ex vivo culture durations while maintaining gene transfer to long-term repopulating cells. We have previously reported long-term gene transfer rates of 12–40% after VSV-G/ lentivirus vector transduction of murine stem cells by targeting unseparated marrow cells after reduced prestimulation and a single 12 hour vector exposure (Kurre et al., Mol. Ther. 2004 Jun;9(6):914–22). We herein report studies showing maintenance of gene transfer efficiency in this model at drastically reduced ex vivo vector exposure times. In initial in vitro experiments we studied cytokine support, vector particle density, and minimum exposure duration requirements for efficient gene transfer to unseparated marrow cells. We determined that fibronectin fragment support was critical in maintaining minimum gene transfer efficiencies, even during brief 1, or 3-hour exposures. In an effort to extend these in vitro findings targeting a mixed leukocyte population and explore the feasibility in vivo, we next performed repopulation experiments in myeloablated murine recipients. Unseparated marrow cells harvested from donor animals were depleted of red blood cells, washed and immediately transduced on fibronectin fragment in the presence of murine stem cell factor. Following a 1 hour exposure to lentivector (VSV-G/RRLsin-cPPThPGK-EGFPwpre), cells were washed repeatedly, resuspended and injected into myeloablated recipients (n=10). Animals showed ready hematopoietic reconstitution and demonstrated average GFP marking of 31% (range: 17–41.2%) in peripheral blood 20 weeks after transplantation. Gene marking in secondary recipients 9 weeks after reconstitution (n=15, 3 recipient animals per donor) persisted at 29% on average (range 14.9–66%). Results also demonstrate transduction of granulocytes, B- and T-lymphocytes, as well as stable long-term GFP expression in primary and secondary animals. Copy number determination by real-time PCR in marrow cells from primary recipients shows an average of 4 proviral copies (range 2.1–8.1) per GFP-expressing cell. Our studies confirm that HIV-derived lentivirus vectors are ideally suited for the transduction of murine long-term repopulating cells. We hypothesize that ultra-short transduction actively preserves stem cell content in the inoculum. Moreover, this protocol represents an ideal platform for subsequent in vivo selection to achieve complete phenotype correction and high-level therapeutic chimerism required for some applications. We anticipate that our strategy may prove particularly useful in situations where the target stem cell quantity is greatly limited and cells are of poor ex vivo viability.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4816-4816
Author(s):  
Stephen L Fischer ◽  
Jacqueline M Fonseca ◽  
Yukang Zhao ◽  
Linda L. Kelley ◽  
Ramasamy Sakthivel

Abstract Abstract 4816 Hematopoietic stem cell (HSC) transplantation has become the standard of care for patients with hematologic cancers, anemia, and a variety of other malignant and non-malignant disorders, with greater than 50,000 such procedures being performed globally each year, according to the Worldwide Network for Blood and Marrow Transplantation. Although mobilized peripheral blood (MPB) has become a preferred source of HSCs for transplants, bone marrow (BM) and umbilical cord blood (UCB) are also frequently utilized. Regardless of source, several groups have reported that grafts containing lower total nucleated cell (TNC) and CD34+ cell doses contribute to delayed engraftment and higher graft failure rate. Therefore, methods to increase the total cell number while maintaining the progenitor phenotype, especially the CD34+ progenitor cells, from individual grafts would have a significant clinical impact. Ex vivo expansion of HSCs prior to transplantation is one approach that offers tremendous promise for increasing cell doses and improving clinical outcomes. In many ex vivo culture systems, HSCs are cultured as a suspension cells and cultured in the presence of various media additives that act to enhance cell proliferation while reducing differentiation. An often-overlooked factor influencing fate decisions is the interaction of HSCs with a substrate. In the natural bone marrow microenvironment, HSCs maintain close contact with a complex network of stromal cells and extracellular matrix, likely indicating that cell-cell and cell-matrix interactions play an important role in maintaining their stem cell phenotype. With the goal of mimicking the bone marrow stem cell niche, Arteriocyte, Inc. has developed a 3-D NANEX nanofiber based cell culture substrate. The functionalized NANEX substrate is designed to provide topographical and substrate-immobilized biochemical cues that act in synergy with media additives to enhance HSC proliferation while maintain the progenitors stem cell phenotype. Here, we present our recent work with the NANEX platform towards comparing and achieving a high yield ex vivo expansion of CD34+ cells from MPB, BM, and UCB. Additionally, through the use of flow cytometry and CFU assays, we quantify and characterize NANEX-expanded cells from each source. Furthermore, we compared NANEX to a variety of commercially available products and demonstrate that NANEX significantly improves expansion and reduces phenotype loss during ex vivo culture. Our data indicates that NANEX technology provides a robust ex vivo expansion of HSCs and, with further GMP and clinical development, offers great potential for clinical applications. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document