scholarly journals Human Cord Blood-Derived AC133+ Progenitor Cells Preserve Endothelial Progenitor Characteristics after Long Term In Vitro Expansion

PLoS ONE ◽  
2010 ◽  
Vol 5 (2) ◽  
pp. e9173 ◽  
Author(s):  
Branislava Janic ◽  
Austin M. Guo ◽  
A. S. M. Iskander ◽  
Nadimpalli Ravi S. Varma ◽  
Alfonso G. Scicli ◽  
...  
PLoS ONE ◽  
2012 ◽  
Vol 7 (5) ◽  
pp. e37577 ◽  
Author(s):  
Branislava Janic ◽  
Kourosh Jafari-Khouzani ◽  
Abbas Babajani-Feremi ◽  
A. S. M. Iskander ◽  
Nadimpalli Ravi S. Varma ◽  
...  

Blood ◽  
2020 ◽  
Vol 136 (21) ◽  
pp. 2410-2415 ◽  
Author(s):  
Peng Hua ◽  
Joanna Hester ◽  
George Adigbli ◽  
Rong Li ◽  
Bethan Psaila ◽  
...  

Abstract Although cytokine-mediated expansion of human hematopoietic stem cells (HSCs) can result in high yields of hematopoietic progenitor cells, this generally occurs at the expense of reduced bone marrow HSC repopulating ability, thereby limiting potential therapeutic applications. Because bromodomain-containing proteins (BCPs) have been demonstrated to regulate mouse HSC self-renewal and stemness, we screened small molecules targeting various BCPs as potential agents for ex vivo expansion of human HSCs. Of 10 compounds tested, only the bromodomain and extra-terminal motif inhibitor CPI203 enhanced the expansion of human cord blood HSCs without losing cell viability in vitro. The expanded cells also demonstrated improved engraftment and repopulation in serial transplantation assays. Transcriptomic and functional studies showed that the expansion of long-term repopulating HSCs was accompanied by synchronized expansion and maturation of megakaryocytes consistent with CPI203-mediated reprogramming of cord blood hematopoietic stem and progenitor cells. This approach may therefore prove beneficial for ex vivo gene editing, for enhanced platelet production, and for the improved usage of cord blood for transplantation research and therapy.


Blood ◽  
2011 ◽  
Vol 117 (18) ◽  
pp. 4773-4777 ◽  
Author(s):  
Hal E. Broxmeyer ◽  
Man-Ryul Lee ◽  
Giao Hangoc ◽  
Scott Cooper ◽  
Nutan Prasain ◽  
...  

Abstract Cryopreservation of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) is crucial for cord blood (CB) banking and transplantation. We evaluated recovery of functional HPC cryopreserved as mononuclear or unseparated cells for up to 23.5 years compared with prefreeze values of the same CB units. Highly efficient recovery (80%-100%) was apparent for granulocyte-macrophage and multipotential hematopoietic progenitors, although some collections had reproducible low recovery. Proliferative potential, response to multiple cytokines, and replating of HPC colonies was extensive. CD34+ cells isolated from CB cryopreserved for up to 21 years had long-term (≥ 6 month) engrafting capability in primary and secondary immunodeficient mice reflecting recovery of long-term repopulating, self-renewing HSCs. We recovered functionally responsive CD4+ and CD8+ T lymphocytes, generated induced pluripotent stem (iPS) cells with differentiation representing all 3 germ cell lineages in vitro and in vivo, and detected high proliferative endothelial colony forming cells, results of relevance to CB biology and banking.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3714-3714
Author(s):  
Joshua A. Wood ◽  
Evan Colletti ◽  
Laura E. Mead ◽  
David A. Ingram ◽  
Christopher D. Porada ◽  
...  

Abstract Endothelial progenitor cells (EPC), whether isolated from the bone marrow (BM), peripheral (PB), or cord blood (CB), represent a promising tool for the development of novel cell therapies. EPC have been shown to contribute to re-endothelialization and neovascularization of damaged tissue, and have been proposed to be some of the primary regulators of tissue regeneration in organs such as the liver. Many studies have looked at the role of EPC in vasculogenic processes, but very few, if any, have focused their efforts on determining the complete differentiative potential of EPC upon transplantation in an experimental model that permits the robust formation of donor-derived tissue-specific cells in the absence of selective pressure to drive differentiation towards a specific phenotype. To this end, CB-derived EPC were obtained as previously described (Ingram et al. Blood:104,2004), transduced with a retroviral vector expressing dsRed, and transplanted (Tx) into 55–60 days old fetal sheep recipients (n=8) at concentrations ranging from 0.5–1.5 × 106cells/fetus. Recipients were then evaluated at 85 days post-transplant for the presence of donor (human)-specific cell types using flow cytometry and confocal microscopy. Using these methods, we found that levels of EPC engraftment in liver, as detected by dsRed expression, correlated directly with the Tx cell dose. Furthermore co-localization of CD31 or vWF was found within the dsRed+ cells. In animals receiving lower cell doses, EPC engrafted throughout the liver at the overall level of 0.12±0.03%; this number doubled in animals that received 2.6 × 106cells. Importantly, there was a preferential distribution of EPC around the vessels, with the EPC comprising 10 to 25% of the cells located around the perivascular areas, with some contributing directly to the endothelial layer of these vessels. Furthermore, expression of Connexin-43 and 45 in engrafted EPC demonstrated that the EPC had not only engrafted, but had also functionally integrated into the developing blood vessels. In addition, co-expression of albumin and alpha-fetoprotein in some of the engrafted EPC suggests that some of these cells may also have contributed to cells with a hepatocyte-like phenotype. Flow cytometric analysis of BM and PB of the transplanted sheep demonstrated that EPC engrafted and proliferated in the BM, with cells expressing CD105 (6.2±2.2) and CD146 (0.6±0.1), and continued to circulate in the PB with cells positive for CD105 (1.4±0.4) and CD146 (0.9±0.2). Of interest is that a CD45 negative aminopeptidase N+ (APN/CD13) population was found in both BM (18±7) and PB (5.6±2). This is particularly interesting, since CD13/APN is a potent regulator of vascular endothelial morphogenesis during angiogenesis. In conclusion, CB derived EPC are able to engraft and proliferate in vivo, integrate into the developing cytoarchitecture, and establish a circulating EPC pool ensuring long-term contribution to ongoing vasculogenesis.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5404-5404
Author(s):  
Eun-Sun Yoo ◽  
Jee-Young Ahn ◽  
KiHwan Kwon ◽  
Soo-Ah Oh ◽  
Moon-Young Choi ◽  
...  

Abstract Background: The identification of circulating endothelial progenitor cells (EPCs) has revolutionized approaches to cell-based therapy for injured and ischemic tissues. Recently, we have demonstrated that there are 2 distinct types of EPCs from UCB having different biologic properties for angiogenic capabilities in vitro and in vivo. In present study, the aim is to directly compare umbilical cord blood (UCB)- and BM-derived late EPC surface phenotypes and in vitro functional capacity. Methods: Mononuclear cells from UCB and BM cultured using EGM-2 medium with VEGF, IGF-1 and FGF for 21 days. Late outgrowing endothelail cells(late OECs) which were in peak growth at third weeks of culture were analyzed for expression of various surface markers by flow cytometry/RT-PCR/IF, tube formation in Matrigel plates, proliferation assay, endothelial colony assay and the role of SDF-1/VEGF on migration. Results: The adherent cells after culture of 7 days exhibited a fibroblast like shape in BM and a cobblestone shaped cells in UCB. Although two sources of OECs were comparable in expression of endothelial and various adhesion molecule markers, BM-derived OECs contained higher proportion of cells expressing smooth muscle cell markers(SMMHC), several adhesion molecule(CD49d, CD62L and VCAM-1), whereas the expression of CXCR-4, PECAM-1 and CD62E and expression of mRNA on endothelial marker genes were higher in UCB-derived OECs. UCB-OECS stained positive for uptake of acetylated low-density lipoprotein and had more migratory ability in the presence of SDF-1 and VEGF compared with BM-OECs. Both sources OECs effectively formed capillary tubes in Matrigel plates. Conclusion: We directly compared OECs derived from UCB and BM and two source of OECs differ in aspect of several adhesion molecule and angiogenic potential in vitro. These difference of UCB render it potentially advantageous for human therapeutic OECs applications for potential applications for a “cell therapy” in the situations on vascular injuries when compared with patients-derived BM.


Sign in / Sign up

Export Citation Format

Share Document