9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Osteoprotegerin Induces CD34 + Differentiation in Endothelial Progenitor Cells

      brief-report

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Endothelial progenitor cells (EPCs) are the main hypothetical cells that could give rise to vessels and in particular one subtype isolated from peripheral or cord bloods: endothelial colony forming cells (ECFCs). These ECFCs are clonogenic precursors committed to endothelial lineage and have robust vasculogenic properties. However, their low number and poor expansion properties when isolated from human adult bloods, currently limit their use as an autologous cell therapy product. We previously reported that osteoprotegerin (OPG), a well-characterized regulator of bone metabolism, contributes to ischemic tissue revascularization, tumor growth in vivo, and potentiates ECFCs proangiogenic properties through the secretion of SDF-1. The current study investigated the role of OPG in ECFCs differentiation and expansion from cord blood CD34 + cells. OPG increased the number of ECFCs after endothelial differentiation of CD34 + cells, enhancing the time of EPCs colonies initial appearance and the growth kinetic of endothelial cell progeny. OPG-exposed ECFCs expressed higher levels of CD34 + compared to control ECFCs. In conclusion, our findings provide novel insights into OPG in regulation of CD34 + progenitor cells. These results give new opportunities for ex vivo expansion of human ECFCs using OPG as a cell culture component for future ECFC product manufacture according to GMP.

          Related collections

          Most cited references30

          • Record: found
          • Abstract: found
          • Article: not found

          Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals.

          The limited vessel-forming capacity of infused endothelial progenitor cells (EPCs) into patients with cardiovascular dysfunction may be related to a misunderstanding of the biologic potential of the cells. EPCs are generally identified by cell surface antigen expression or counting in a commercially available kit that identifies "endothelial cell colony-forming units" (CFU-ECs). However, the origin, proliferative potential, and differentiation capacity of CFU-ECs is controversial. In contrast, other EPCs with blood vessel-forming ability, termed endothelial colony-forming cells (ECFCs), have been isolated from human peripheral blood. We compared the function of CFU-ECs and ECFCs and determined that CFU-ECs are derived from the hematopoietic system using progenitor assays, and analysis of donor cells from polycythemia vera patients harboring a Janus kinase 2 V617F mutation in hematopoietic stem cell clones. Further, CFU-ECs possess myeloid progenitor cell activity, differentiate into phagocytic macrophages, and fail to form perfused vessels in vivo. In contrast, ECFCs are clonally distinct from CFU-ECs, display robust proliferative potential, and form perfused vessels in vivo. Thus, these studies establish that CFU-ECs are not EPCs and the role of these cells in angiogenesis must be re-examined prior to further clinical trials, whereas ECFCs may serve as a potential therapy for vascular regeneration.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            In vivo vasculogenic potential of human blood-derived endothelial progenitor cells.

            Vascularization of tissues is a major challenge of tissue engineering (TE). We hypothesize that blood-derived endothelial progenitor cells (EPCs) have the required proliferative and vasculogenic activity to create vascular networks in vivo. To test this, EPCs isolated from human umbilical cord blood or from adult peripheral blood, and human saphenous vein smooth muscle cells (HSVSMCs) as a source of perivascular cells, were combined in Matrigel and implanted subcutaneously into immunodeficient mice. Evaluation of implants at one week revealed an extensive network of human-specific lumenal structures containing erythrocytes, indicating formation of functional anastomoses with the host vasculature. Quantitative analyses showed the microvessel density was significantly superior to that generated by human dermal microvascular endothelial cells (HDMECs) but similar to that generated by human umbilical vein endothelial cells (HUVECs). We also found that as EPCs were expanded in culture, their morphology, growth kinetics, and proliferative responses toward angiogenic factors progressively resembled those of HDMECs, indicating a process of in vitro maturation. This maturation correlated with a decrease in the degree of vascularization in vivo, which could be compensated for by increasing the number of EPCs seeded into the implants. Our findings strongly support the use of human EPCs to form vascular networks in engineered organs and tissues.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Differentiation of human pluripotent stem cells to cells similar to cord-blood endothelial colony-forming cells.

              The ability to differentiate human pluripotent stem cells into endothelial cells with properties of cord-blood endothelial colony-forming cells (CB-ECFCs) may enable the derivation of clinically relevant numbers of highly proliferative blood vessel-forming cells to restore endothelial function in patients with vascular disease. We describe a protocol to convert human induced pluripotent stem cells (hiPSCs) or embryonic stem cells (hESCs) into cells similar to CB-ECFCs at an efficiency of >10(8) ECFCs produced from each starting pluripotent stem cell. The CB-ECFC-like cells display a stable endothelial phenotype with high clonal proliferative potential and the capacity to form human vessels in mice and to repair the ischemic mouse retina and limb, and they lack teratoma formation potential. We identify Neuropilin-1 (NRP-1)-mediated activation of KDR signaling through VEGF165 as a critical mechanism for the emergence and maintenance of CB-ECFC-like cells.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Med (Lausanne)
                Front Med (Lausanne)
                Front. Med.
                Frontiers in Medicine
                Frontiers Media S.A.
                2296-858X
                27 November 2018
                2018
                : 5
                : 331
                Affiliations
                [1] 1Inserm, UMR_S1140, Faculty of Pharmacy, Université Paris Descartes, Sorbonne Paris Cité , Paris, France
                [2] 2Inserm, UMR_S1232, CRCINA, Institut de Cancérologie de l'Ouest, Université Nantes-Angers-Le Mans , Nantes, France
                [3] 3AP-HP, Hematology Department, European Georges Pompidou Hospital , Paris, France
                Author notes

                Edited by: Ondrej Viklicky, Institute for Clinical and Experimental Medicine (IKEM), Czechia

                Reviewed by: Marian Klinger, Opole University, Poland; Tomomi Toubai, Yamagata University, Japan

                *Correspondence: Catherine Boisson-Vidal catherine.boisson-vidal@ 123456parisdescartes.fr

                This article was submitted to Translational Medicine, a section of the journal Frontiers in Medicine

                Article
                10.3389/fmed.2018.00331
                6277572
                30538990
                52077662-7fd8-4b81-8d4a-8e4578d95f86
                Copyright © 2018 Boisson-Vidal, Benslimane-Ahmim, Lokajczyk, Heymann and Smadja.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 13 July 2018
                : 12 November 2018
                Page count
                Figures: 2, Tables: 0, Equations: 0, References: 35, Pages: 7, Words: 4087
                Categories
                Medicine
                Brief Research Report

                osteoprotegerin,endothelial progenitor cells,endothelial-colony forming cells,cd34+ cells,proliferation

                Comments

                Comment on this article