scholarly journals Identification of Putative Markers That Predict the In Vitro Senescence of Mesenchymal Progenitor Cells

Cells ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 1301
Author(s):  
Eun-Young Shin ◽  
Yeo-Joon Yoon ◽  
Jeoung Eun Lee ◽  
Sung Han Shim ◽  
Gene Hong Park ◽  
...  

Mesenchymal progenitor cells (MPCs) are a promising cell source for regenerative medicine because of their immunomodulatory properties, anti-inflammatory molecule secretion, and replacement of damaged cells. Despite these advantages, heterogeneity in functional potential and limited proliferation capacity of MPCs, as well as the lack of suitable markers for product potency, hamper the development of large-scale manufacturing processes of MPCs. Therefore, there is a sustained need to develop highly proliferative and standardized MPCs in vitro and find suitable functional markers for measuring product potency. In this study, three lines of pluripotent stem cell (PSC)-derived MPCs with high proliferative ability were established and compared with bone-marrow-derived MPCs using proliferation assays and microarrays. A total of six genes were significantly overexpressed (>10-fold) in the highest proliferative MPC line (CHA-hNT5-MPCs) and validated by qRT-PCR. However, only two of the genes (MYOCD and ODZ2) demonstrated a significant correlation with MPC senescence in vitro. Our study provides new gene markers for predicting replicative senescence and the available quantity of MPCs but may also help to guide the development of new standard criteria for manufacturing.

2014 ◽  
Vol 29 (4) ◽  
pp. 457-469 ◽  
Author(s):  
Federica Riva ◽  
Claudia Omes ◽  
Roberto Bassani ◽  
Rossella E Nappi ◽  
Giuliano Mazzini ◽  
...  

2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Sadaf Vahdat ◽  
Sara Pahlavan ◽  
Elena Mahmoudi ◽  
Maryam Barekat ◽  
Hassan Ansari ◽  
...  

Abstract Cardiovascular progenitor cells (CPCs) derived from human pluripotent stem cells (hPSCs) are proposed to be invaluable cell sources for experimental and clinical studies. This wide range of applications necessitates large-scale production of CPCs in an in vitro culture system, which enables both expansion and maintenance of these cells. In this study, we aimed to develop a defined and efficient culture medium that uses signaling factors for large-scale expansion of early CPCs, called cardiogenic mesodermal cells (CMCs), which were derived from hPSCs. Chemical screening resulted in a medium that contained a reproducible combination of three factors (A83-01, bFGF, and CHIR99021) that generated 1014 CMCs after 10 passages without the propensity for tumorigenicity. Expanded CMCs retained their gene expression pattern, chromosomal stability, and differentiation tendency through several passages and showed both the safety and possible cardio-protective potentials when transplanted into the infarcted rat myocardium. These CMCs were efficiently cryopreserved for an extended period of time. This culture medium could be used for both adherent and suspension culture conditions, for which the latter is required for large-scale CMC production. Taken together, hPSC-derived CMCs exhibited self-renewal capacity in our simple, reproducible, and defined medium. These cells might ultimately be potential, promising cell sources for cardiovascular studies.


2009 ◽  
Vol 27 (2) ◽  
pp. 208-215 ◽  
Author(s):  
Takashi Iwakura ◽  
Masahiko Miwa ◽  
Yoshitada Sakai ◽  
Takahiro Niikura ◽  
Sang Yang Lee ◽  
...  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4105-4105
Author(s):  
Brenton Short ◽  
Emer Clarke ◽  
Terry Thomas ◽  
Allen C. Eaves ◽  
Albertus W. Wognum ◽  
...  

Abstract Adult mammalian bone marrow (BM) contains at least two distinct stem cell populations; the stem cells of the hemopoietic lineage and a second population termed mesenchymal stem cells (MSC) whose function is to maintain the non-hemopoietic BM elements as well as skeletal homeostasis. MSC have been implicated as potential targets in a range of cellular therapies for treatment of defects of both the hemopoietic and skeletal systems, and as vehicles for gene therapy. In order to evaluate the potential of these cells in various therapies, a pre-clinical animal model in which both the biology and potential therapeutic applications of these cells can be assessed is of fundamental importance. The goal of the current study was to develop a robust and reproducible method for the isolation of MSC from murine hemopoietic tissues. Tibiae and femurs harvested from C57BL6/J mice were gently crushed with a pestle to release the marrow. The bone fragments were subsequently cut into small pieces with a scalpel and digested in a solution containing 3mg/ml Type I collagenase to yield a population of compact bone (CB) derived cells. Mesenchymal progenitor cells (MPC) were detected using an in vitro assay for fibroblast-colony forming cells (CFU-F). CB cells were plated at 1000 or 5000 cells per cm2 in complete MesenCult™ medium for 12 days, after which CFU-F-derived colonies were enumerated. We show that CFU-F are present at a significantly higher frequency in mouse CB than in the BM (433±225 vs 11.7±3.5 colonies/106 cells respectively, n=3). Based on these data we developed a simple and robust immunomagnetic selection method to highly enrich MPC from mouse CB by depleting essentially all nucleated hemopoietic cells (CD45+) and red blood cells (Ter119+) using magnetic particles and antibodies to CD45 and Ter119, respectively. Target CD45−Ter119− cells initially comprised 1.1±0.5% (n=9) of the total CB fraction as assayed by FACS. Following depletion, CD45−Ter119− cells comprised 74.5±16% (n=6) of the cells and were enriched 205 fold for CFU-F compared to the starting population, with a CFU-F frequency of 1 per 11 cells plated, and a total CFU-F recovery of 57.9 ± 18.5%. Analysis of CD45−Ter119−Sca-1+ cells, a phenotype previously shown to enrich for MPC, revealed that these cells were enriched 50 fold following depletion, from 0.53±0.5 to 26.5±8.23% (n=3). The enriched MPCs cultured at low O2 tension were devoid of hemopoietic contaminants at passage 1 and 2 as shown by lack of CD45, Ter119 and CD11b expression. The cultured CB-derived MPCs were capable of extensive in vitro proliferation and maintained the ability to differentiate into cells of the osteogenic, adipogenic and chondrogenic lineages. Furthermore, irradiated cultured mesenchymal cells supported long-term culture-initiating cells (LTC-IC) in 4-week cultures of Sca-1+ BM cells under limiting dilution conditions, at frequencies similar to those detected using irradiated primary BM feeders (i.e. 1 per 1600). These data provide a rapid, reproducible method by which multipotent mesenchymal cells devoid of contaminating hemopoietic cells can be readily obtained from limited numbers of mice to study the biology of MSC as well as the use of these cells as therapeutic agents in a preclinical animal model.


Blood ◽  
2009 ◽  
Vol 113 (26) ◽  
pp. 6716-6725 ◽  
Author(s):  
Andreas Reinisch ◽  
Nicole A. Hofmann ◽  
Anna C. Obenauf ◽  
Karl Kashofer ◽  
Eva Rohde ◽  
...  

Abstract Endothelial progenitor cells are critically involved in essential biologic processes, such as vascular homeostasis, regeneration, and tumor angiogenesis. Endothelial colony–forming cells (ECFCs) are endothelial progenitor cells with robust proliferative potential. Their profound vessel-forming capacity makes them a promising tool for innovative experimental, diagnostic, and therapeutic strategies. Efficient and safe methods for their isolation and expansion are presently lacking. Based on the previously established efficacy of animal serum–free large-scale clinical-grade propagation of mesenchymal stromal cells, we hypothesized that endothelial lineage cells may also be propagated efficiently following a comparable strategy. Here we demonstrate that human ECFCs can be recovered directly from unmanipulated whole blood. A novel large-scale animal protein-free humanized expansion strategy preserves the progenitor hierarchy with sustained proliferation potential of more than 30 population doublings. By applying large-scale propagated ECFCs in various test systems, we observed vascular networks in vitro and perfused vessels in vivo. After large-scale expansion and cryopreservation phenotype, function, proliferation, and genomic stability were maintained. For the first time, proliferative, functional, and storable ECFCs propagated under humanized conditions can be explored in terms of their therapeutic applicability and risk profile.


Sign in / Sign up

Export Citation Format

Share Document