Evidence That Human Very Small Embryonic-Like Stem Cells (VSELs) Are Mobilized by G-CSF Into Peripheral Blood: A Novel Strategy to Obtain Human Pluripotent Stem Cells for Regenerative Medicine.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1474-1474
Author(s):  
Satish Medicetty ◽  
Mariusz Z Ratajczak ◽  
Magdalena J Kucia ◽  
Ewa K. Zuba-Surma ◽  
Izabela Klich ◽  
...  

Abstract Abstract 1474 Poster Board I-497 We previously demonstrated that human cord blood contains a population of small (smaller in size than erythrocytes) CXCR4+CD133+CD34+SSEA-4+Oct-4+lin−CD45− cells (Leukemia 2007:21;297-303) and that these cells are mobilized into peripheral blood during tissue organ damage as seen for example in heart infarct (J. Am. Coll. Cardiol., 2009:53;1-9.) or stroke (Stroke. 2009:40;1237.). Similar cells were also reported in murine organs, and more importantly we described that these cells may differentiate in vitro into cells from all three germ layers (Leukemia 2006:20;857–869). To explore the possibility that human VSELs could become a source of pluripotent stem cells in regenerative medicine, our goal was to develop an efficient strategy to isolate these cells from adult patients. To test if VSELs similarly to their murine counterparts could be mobilized into peripheral blood after granulocyte colony stimulating factor (G-CSF) injection (Stem Cells 2008:26;2083-2092), we enrolled a group of young healthy donors who were mobilized for two consecutive days using G-CSF (480 μg/day subcutaneously). On the third day nucleated cells (TNC) were collected by apheresis. We evaluated number of VSELs in peripheral blood (PB) samples before and after G-CSF mobilization as well as the final number in the apheresis product. At least 1 million of TNC were acquired and analyzed by FACS Diva software. Three different fractions of non-hematopoietic stem cells enriched for VSELs (Lin−/CD45−/CD133+, Lin−/CD45−/CD34+, Lin−/CD45−/CXCR4+) as well as their CD45 positive hematopoietic counterparts were analyzed. The absolute numbers of cells from each population, contained in 1 μL of sample, were computed based on percent content of each population and TNC count for each individual sample. Results show that after G-CSF mobilization, human peripheral blood contains a population of lin− CD45− mononuclear cells that express CXCR4, CD34 and CD133 antigens. These lin− CD45− CXCR4+ CD133+ CD34+ cells are highly enriched for mRNA for intra-nuclear pluripotent embryonic transcription factors such as Oct-4, Sox2 and Nanog. More importantly we found that Oct-4 was expressed in nuclei of mobilized VSELs and that these cells also express the cell surface marker SSEA-4, the early embryonic glycolipid antigen commonly used as a marker for undifferentiated pluripotent human embryonic stem cells. We observed that these adult peripheral blood-derived VSELs are slightly larger than their counterparts identified in adult murine bone marrow, but are still very small. In addition, they also possess large nuclei containing embryonic-type unorganized euchromatin. Before G-CSF mobilization only very few VSELs were detectable in peripheral blood, whereas following G-CSF induced mobilization there was a very significant increase with in excess of 106 VSELs present in the apheresis product representing less than 0.01% of TNC. We postulate that while VSELs are relatively rare cells, they are mobilized into peripheral blood and that G-CSF induced mobilization could become a novel strategy to obtain human pluripotent stem cells for regenerative medicine. Disclosures: Medicetty: NeoStem Inc: Employment, Equity Ownership. Marasco: NeoStem Inc: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Rodgerson: NeoStem Inc: Employment, Equity Ownership.

2021 ◽  
Author(s):  
Lea Flippe ◽  
Anne Gaignerie ◽  
Celine Serazin ◽  
Olivier Baron ◽  
Xavier Saulquin ◽  
...  

Immunotherapy using primary T cells has revolutionized medical care in some pathologies in recent years but limitations associated to challenging cell genome edition, insufficient cell number production, the use of only autologous cells and lack of product standardization have limited its uses in the clinic. The alternative use of T cells generated in vitro from human pluripotent stem cells (hPSCs) offers great advantages by providing a self-renewing source of T cells that can be readily genetically modified and facilitate the use of standardized universal off-the-shelf allogeneic cell products and rapid clinic access. However, despite their potential, the feasibility and functionality of T-cells differentiated from hPSCs needs better comprehension before moving to the clinic. In this study, we generated human induced pluripotent stem cells from T-cells (T-iPSCs) allowing preservation of already recombined TCR, with the same properties as human embryonic stem cells (hESCs). Based on these cells, we differentiated with high efficiency hematopoietic progenitor stem cells (HPSCs), capable of self-renewal and differentiation into any cell blood type, and then DN3a thymic progenitors from several T-iPSC lines. To better comprehend differentiation, we analyzed the transcriptomic profiles of the different cell types and demonstrated that HPSCs differentiated from hiPSCs had a very similar profile to cord blood hematopoietic stem cells (HSCs). Furthermore, differentiated T-cell progenitors had a similar profile to thymocytes at the DN3a stage of thymic lymphopoiesis. Therefore, with this approach, we were able to regenerate precursors of therapeutic human T cells to potentially treat a wide number of diseases.


2019 ◽  
Vol 20 (7) ◽  
pp. 1737 ◽  
Author(s):  
Stéphane Flamant ◽  
Jean-Claude Chomel ◽  
Christophe Desterke ◽  
Olivier Féraud ◽  
Emilie Gobbo ◽  
...  

Although human pluripotent stem cells (hPSCs) can theoretically differentiate into any cell type, their ability to produce hematopoietic cells is highly variable from one cell line to another. The underlying mechanisms of this heterogeneity are not clearly understood. Here, using a whole miRNome analysis approach in hPSCs, we discovered that their hematopoietic competency was associated with the expression of several miRNAs and conversely correlated to that of miR-206 specifically. Lentiviral-based miR-206 ectopic expression in H1 hematopoietic competent embryonic stem (ES) cells markedly impaired their differentiation toward the blood lineage. Integrative bioinformatics identified a potential miR-206 target gene network which included hematopoietic master regulators RUNX1 and TAL1. This work sheds light on the critical role of miR-206 in the generation of blood cells off hPSCs. Our results pave the way for future genetic manipulation of hPSCs aimed at increasing their blood regenerative potential and designing better protocols for the generation of bona fide hPSC-derived hematopoietic stem cells.


Blood ◽  
2009 ◽  
Vol 114 (17) ◽  
pp. 3513-3523 ◽  
Author(s):  
Dan S. Kaufman

Abstract Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) provide remarkable cellular platforms to better understand human hematopoiesis and to develop clinically applicable hematopoietic cell–based therapies. Over the past decade, hESCs have been used to characterize molecular and cellular mechanisms underpinning the differentiation of hematopoietic progenitors and mature, functional hematopoietic cells. These advances are now poised to lead to clinical translation of hESC- and iPSC-derived hematopoietic cells for novel therapies in the next few years. On the basis of areas of recent success, initial clinical use of hematopoietic cells derived from human pluripotent stem cells will probably be in the areas of transfusion therapies (erythrocytes and platelets) and immune therapies (natural killer cells). In contrast, efficient development and isolation of hematopoietic stem cells capable of long-term, multilineage engraftment still remains a significant challenge. Technical, safety, and regulatory concerns related to clinical applications of human PSCs must be appropriately addressed. However, proper consideration of these issues should facilitate and not inhibit clinical translation of new therapies. This review outlines the current status of hematopoietic cell development and what obstacles must be surmounted to bring hematopoietic cell therapies from human PSCs from “bench to bedside.”


2020 ◽  
Vol 15 (4) ◽  
pp. 301-307 ◽  
Author(s):  
Gaifang Wang ◽  
Maryam Farzaneh

Primary Ovarian Insufficiency (POI) is one of the main diseases causing female infertility that occurs in about 1% of women between 30-40 years of age. There are few effective methods for the treatment of women with POI. In the past few years, stem cell-based therapy as one of the most highly investigated new therapies has emerged as a promising strategy for the treatment of POI. Human pluripotent stem cells (hPSCs) can self-renew indefinitely and differentiate into any type of cell. Human Embryonic Stem Cells (hESCs) as a type of pluripotent stem cells are the most powerful candidate for the treatment of POI. Human-induced Pluripotent Stem Cells (hiPSCs) are derived from adult somatic cells by the treatment with exogenous defined factors to create an embryonic-like pluripotent state. Both hiPSCs and hESCs can proliferate and give rise to ectodermal, mesodermal, endodermal, and germ cell lineages. After ovarian stimulation, the number of available oocytes is limited and the yield of total oocytes with high quality is low. Therefore, a robust and reproducible in-vitro culture system that supports the differentiation of human oocytes from PSCs is necessary. Very few studies have focused on the derivation of oocyte-like cells from hiPSCs and the details of hPSCs differentiation into oocytes have not been fully investigated. Therefore, in this review, we focus on the differentiation potential of hPSCs into human oocyte-like cells.


Cells ◽  
2020 ◽  
Vol 9 (5) ◽  
pp. 1078
Author(s):  
Tae Won Ha ◽  
Ji Hun Jeong ◽  
HyeonSeok Shin ◽  
Hyun Kyu Kim ◽  
Jeong Suk Im ◽  
...  

Human pluripotent stem cells (hPSCs), such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), have a well-orchestrated program for differentiation and self-renewal. However, the structural features of unique proteostatic-maintaining mechanisms in hPSCs and their features, distinct from those of differentiated cells, in response to cellular stress remain unclear. We evaluated and compared the morphological features and stress response of hPSCs and fibroblasts. Compared to fibroblasts, electron microscopy showed simpler/fewer structures with fewer networks in the endoplasmic reticulum (ER) of hPSCs, as well as lower expression of ER-related genes according to meta-analysis. As hPSCs contain low levels of binding immunoglobulin protein (BiP), an ER chaperone, thapsigargin treatment sharply increased the gene expression of the unfolded protein response. Thus, hPSCs with decreased chaperone function reacted sensitively to ER stress and entered apoptosis faster than fibroblasts. Such ER stress-induced apoptotic processes were abolished by tauroursodeoxycholic acid, an ER-stress reliever. Hence, our results revealed that as PSCs have an underdeveloped structure and express fewer BiP chaperone proteins than somatic cells, they are more susceptible to ER stress-induced apoptosis in response to stress.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Ping Zhou ◽  
Jia-Min Shi ◽  
Jing-E Song ◽  
Yu Han ◽  
Hong-Jiao Li ◽  
...  

Abstract Background Derivation of osteoblast-like cells from human pluripotent stem cells (hPSCs) is a popular topic in bone tissue engineering. Although many improvements have been achieved, the low induction efficiency because of spontaneous differentiation hampers their applications. To solve this problem, a detailed understanding of the osteogenic differentiation process of hPSCs is urgently needed. Methods Monolayer cultured human embryonic stem cells and human-induced pluripotent stem cells were differentiated in commonly applied serum-containing osteogenic medium for 35 days. In addition to traditional assays such as cell viability detection, reverse transcription-polymerase chain reaction, immunofluorescence, and alizarin red staining, we also applied studies of cell counting, cell telomerase activity, and flow cytometry as essential indicators to analyse the cell type changes in each week. Results The population of differentiated cells was quite heterogeneous throughout the 35 days of induction. Then, cell telomerase activity and cell cycle analyses have value in evaluating the cell type and tumourigenicity of the obtained cells. Finally, a dynamic map was made to integrate the analysis of these results during osteogenic differentiation of hPSCs, and the cell types at defined stages were concluded. Conclusions Our results lay the foundation to improve the in vitro osteogenic differentiation efficiency of hPSCs by supplementing with functional compounds at the desired stage, and then establishing a stepwise induction system in the future.


2015 ◽  
Vol 2015 ◽  
pp. 1-19 ◽  
Author(s):  
J.-F. Stoltz ◽  
N. de Isla ◽  
Y. P. Li ◽  
D. Bensoussan ◽  
L. Zhang ◽  
...  

Since the 1960s and the therapeutic use of hematopoietic stem cells of bone marrow origin, there has been an increasing interest in the study of undifferentiated progenitors that have the ability to proliferate and differentiate into various tissues. Stem cells (SC) with different potency can be isolated and characterised. Despite the promise of embryonic stem cells, in many cases, adult or even fetal stem cells provide a more interesting approach for clinical applications. It is undeniable that mesenchymal stem cells (MSC) from bone marrow, adipose tissue, or Wharton’s Jelly are of potential interest for clinical applications in regenerative medicine because they are easily available without ethical problems for their uses. During the last 10 years, these multipotent cells have generated considerable interest and have particularly been shown to escape to allogeneic immune response and be capable of immunomodulatory activity. These properties may be of a great interest for regenerative medicine. Different clinical applications are under study (cardiac insufficiency, atherosclerosis, stroke, bone and cartilage deterioration, diabetes, urology, liver, ophthalmology, and organ’s reconstruction). This review focuses mainly on tissue and organ regeneration using SC and in particular MSC.


Author(s):  
Mayuko Kano ◽  
Hidetaka Suga ◽  
Hiroshi Arima

Abstract The hypothalamus and pituitary have been identified to play essential roles in maintaining homeostasis. Various diseases can disrupt the functions of these systems, which can often result in serious lifelong symptoms. The current treatment for hypopituitarism involves hormone replacement therapy. However, exogenous drug administration cannot mimic the physiological changes that are a result of hormone requirements. Therefore, patients are at a high risk of severe hormone deficiency, including adrenal crisis. Pluripotent stem cells (PSCs) self-proliferate and differentiate into all types of cells. The generation of endocrine tissues from PSCs has been considered as another new treatment for hypopituitarism. Our colleagues established a three-dimensional culture method for embryonic stem cells (ESCs). In this culture, the ESC-derived aggregates exhibit self-organization and spontaneous formation of highly ordered patterning. Recent results have shown that strict removal of exogenous patterning factors during early differentiation efficiently induces rostral hypothalamic progenitors from mouse ESCs. These hypothalamic progenitors generate vasopressinergic neurons, which release neuropeptides upon exogenous stimulation. Subsequently, we reported adenohypophysis tissue self-formation in three-dimensional cultures of mouse ESCs. The ESCs were found to differentiate into both non-neural oral ectoderm and hypothalamic neuroectoderm in adjacent layers. Interactions between the two tissues appear to be critically important for in vitro induction of a Rathke's pouch-like developing embryo. Various endocrine cells were differentiated from non-neural ectoderm. The induced corticotrophs efficiently secreted adrenocorticotropic hormone when engrafted in vivo, which rescued hypopituitary hosts. For future regenerative medicine, generation of hypothalamic and pituitary tissues from human PSCs is necessary. We and other groups succeeded in establishing a differentiation method with the use of human PSCs. Researchers could use these methods for models of human diseases to elucidate disease pathology or screen potential therapeutics.


Sign in / Sign up

Export Citation Format

Share Document