Endothelial Progenitor Cells (EPCs) Isolated from the Rhesus Macaque but Not the Mouse Are Phenotypically and Functionally Similar to Human EPCs.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3910-3910
Author(s):  
David A. Ingram ◽  
Laura E. Mead ◽  
Daniel B. Moore ◽  
Theresa Krier ◽  
Ann Farese ◽  
...  

Abstract We recently identified a novel hierarchy of human endothelial progenitor cells (EPCs), which are functionally defined by their proliferative and clonogenic potential (Blood, 2004). Emerging evidence suggests that EPCs may be used as angiogenic therapies, or as biomarkers to assess cardiovascular disease risk. Thus, identification of animal models, which phenocopy the human EPC hierarchy, is an important priority for preclinical testing of experimental therapeutics. Given the importance of the Rhesus Macaque as a preclinical model, we tested whether EPCs could be isolated from the peripheral blood of the Rhesus Macaque and compared to EPCs isolated from human adult peripheral blood. Mononuclear cells were isolated from 20 ml of Rhesus peripheral blood and cultured in EGM-2 medium, which promotes the formation of EPC colonies. After 7 days in culture, we identified approximately 20 endothelial cell colonies (n=9), which appeared identical to human EPC colonies. We subcultured the endothelial cell colonies into monolayers for immunophenotyping and functional analysis. Endothelial cells (ECs) derived from the Rhesus EPC colonies formed vessels in matrigel, and demonstrated uptake of acetylated LDL, which are characteristics of ECs. Similar to ECs derived from human EPCs, Rhesus ECs expressed the endothelial cell antigens, CD31, CD144, CD105, CD146, and Flk1. Importantly, Rhesus ECs did not express the hematopoietic cell specific antigens, CD45 and CD14. Similar to ECs derived from human peripheral blood EPC colonies, Rhesus ECs could be serially passaged for at least 40 population doublings without signs of cellular senescence. A hallmark of stem and progenitor cells is their ability to proliferate and give rise to functional progeny. Analogous to a paradigm established in the hematopoietic cell system, we recently developed a single cell deposition assay to reproducibly identify the following human EPCs: (1) high proliferative potential - endothelial colony forming cells (HPP-ECFC), which form macroscopic colonies that form secondary and tertiary colonies upon replating, (2) low proliferative potential - endothelial colony forming cells (LPP-ECFC), which form colonies greater than 50 cells, but do not form secondary colonies upon replating, (3) endothelial cell clusters (EC-clusters) that contain less than 50 cells, and (4) mature terminally differentiated endothelial cells (EC), which do not divide (Blood, 2004). To determine whether these different populations of EPCs could be identified in the ECs derived from Rhesus EPCs, we performed single cells deposition assays on 1,000 cells. All types of EPCs could be identified in the Rhesus ECs (Table I). Further, ECs derived from the Rhesus EPCs rapidly form chimeric vessels with human ECs derived from adult blood, implying that the molecular mechanisms critical for vessel formation are conserved between the two species. Finally, while the murine model is an animal model widely used for studying EPCs, a similar hierarchy of EPCs could not be established from the peripheral blood of mice. Thus, given the diversity of therapeutic applications of EPCs for treating a variety of human diseases, these studies establish the Rhesus Macaque as an important preclinical model. Percent of 1,000 Single Cells Plated Mature EC EC-Cluster LPP-ECFC HPP-ECFC Rhesus ECs 85.8±2.1 4.2±1.1 7.8±0.5 1.3±0.5 Human ECs 80.8±9.6 8.6±1.4 12.4±8.1 0.2±0.2

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3912-3912
Author(s):  
Matthew M. Harkenrider ◽  
Scott A. Johnson ◽  
Laura E. Mead ◽  
David A. Ingram ◽  
Mervin C. Yoder

Abstract Endothelial cell replication in large and small vessels is generally thought to occur at a rate of 0.1–0.6% daily. Despite this low level of cell turnover, endothelial cells derived from a variety of bovine vessels display vigorous patterns of proliferation in vitro. This apparent paradox has not been resolved to date. We have recently determined that human endothelial cells are derived through a process of endopoiesis via a hierarchy of endothelial progenitor cells (EPCs) (Blood, 2004). We have developed a single cell proliferation assay that has resolved endopoiesis into distinct stages of progenitor cell development: 1) high proliferative potential-endothelial colony forming cells (HPP-ECFC; 2001-> 10,000 cells/colony) that replate into secondary and tertiary HPP-ECFC, 2) low proliferative potential-endothelial colony forming cells (LPP-ECFC; 51–2,000 cells/colony) that form colonies greater than 50 cells but fail to replate into LPP-ECFC, 3) endothelial clusters (EC-clusters; 2–50 cells/colony) that contain fewer than 50 cells, and 4) mature differentiated endothelial cells that are non-proliferative. We hypothesized that the proliferative behavior of the bovine vessel-derived endothelial cells was due to the presence of EPCs. We purchased bovine aortic endothelial cells (BAEC), bovine pulmonary artery endothelial cells (BPAEC), and bovine coronary artery endothelial cells (BCAEC) from a commercial vendor and cultured the cells as recommended. As predicted, the endothelial cells displayed a cobblestone morphology and ingested acetylated low density lipoprotein consistent with an endothelial phenotype. We initially plated the monolayer of cells of each type at 10, 25, or 100 cells per collagen I coated 6-well tissue culture wells and determined that cells from each artery gave rise to heterogenous colony sizes with different growth potentials during a 7 day culture. We then utilized flow cytometry to single cell sort the endothelial cells of each arterial type and determined the number of cells that divided in a 14 day culture. As depicted in the TABLE, the entire hierarchy of EPCs (similar to that determined for human adult peripheral blood and umbilical cord blood) is present in the endothelial cells isolated from the bovine vessels. Of interest, our preliminary data indicate that the frequency of the most proliferative progenitors (HPP-ECFC) is higher in the BAEC than the BPAEC or BCAEC samples. These data provide a new conceptual framework for understanding the mechanisms of endothelial replacement and/or repair of aged or damaged endothelial cells. While EPCs clearly circulate, they also engraft and reside in the vessel wall. We speculate that it is the presence of these EPCs that accounts for the ability of isolated BAEC, BPAEC, and BCAEC cells to proliferate ex vivo. Single Cell Sort Colony Distributions Cell Line BAEC-1 % BAEC-2 % BCAEC % BPAEC % Mature EC 31.33 39.33 56.67 53.67 EC-clusters 2.00 2.33 10.00 5.00 LPP-ECFC 5.00 9.00 12.00 11.00 HPP-ECFC 61.67 49.33 21.33 30.33 Total colonies 68.67 60.67 43.33 46.33


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2612-2612
Author(s):  
David A. Ingram ◽  
Laura E. Mead ◽  
Wayne Woodard ◽  
Amy Fenoglio ◽  
Brian Murphy ◽  
...  

Abstract Endothelial progenitor cells (EPCs) can be isolated from adult peripheral and umbilical cord blood. EPCs are thought to originate from bone marrow, circulate in peripheral blood, and migrate to sites of angiogenesis. However, the number of circulating EPCs in peripheral blood is remarkably low, and recent genetic studies show that the contribution of bone marrow derived EPCs to newly formed vascular networks is minimal. Further, while endothelial cells (ECs) derived from vessel walls are widely considered to be differentiated mature ECs, these cells retain extensive proliferative potential and can be passaged for at least 40 population doubling in vitro. Based on these observations, we tested whether EPCs potentially reside in vessel walls utilizing a newly developed single cell deposition assay (Blood, 2004). Analogous to a paradigm established in the hematopoietic cell system, we can utilize this assay to reproducibly identify the following EPCs: (1) high proliferative potential - endothelial colony forming cells (HPP-ECFC), which form macroscopic colonies that form secondary and tertiary colonies upon replating, (2) low proliferative potential - endothelial colony forming cells (LPP-ECFC), which form colonies greater than 50 cells, but do not form secondary colonies upon replating, (3) endothelial cell clusters (EC-clusters) that contain less than 50 cells, and (4) mature terminally differentiated endothelial cells (EC), which do not divide. Utilizing this assay, we compared the clonogenic potential of 1000 single adult human dermal microvascular endothelial cells (HMVECds), human umbilical vein endothelial cells (HUVECs), human umbilical artery endothelial cells (HUAECs), human coronary artery endothelial cells (HCAECs), and human aortic endothelial cells (HAECs) to the potential of adult peripheral and umbilical cord blood derived EPCs. We conducted four independent experiments. Remarkably, we demonstrate that a complete hierarchy of EPCs can be identified in EC populations derived from every vessel wall tested (Table I and n=4). Further, we show that ECs derived from each vessel wall cell population tested contain more proliferative EPCs (LPP-ECFCs and HPP-ECFCs) compared to EPCs derived from adult peripheral blood. Percent of 1,000 Single Cells Plated Mature EC EC-Cluster LPP-ECFC HPP-ECFC HUVEC 42±6 18±2 29±9 11±5 HAEC 37±3 23±8 21±4 20±6 HMVECd 65±9 21±6 12±4 2±0.6 HCAEC 46±2 18±2 20±2 16±2 HUAEC 41±1 10±1 27±4 21±2 Adult EPC 81±9 9±1 12±8 0.2±0.2 Cord EPC 50±20 7±2 20±10 23±9 Thus, this study provides evidence that a diversity of EPCs exists in human vessels and provides a new conceptual framework for determining both the origin and function of EPCs in maintaining vessel integrity and contributing to new sites of angiogenesis.


2009 ◽  
Vol 106 (1) ◽  
pp. 203-211 ◽  
Author(s):  
Syotaro Obi ◽  
Kimiko Yamamoto ◽  
Nobutaka Shimizu ◽  
Shinichiro Kumagaya ◽  
Tomomi Masumura ◽  
...  

Endothelial progenitor cells (EPCs) are mobilized from bone marrow to peripheral blood and contribute to angiogenesis in tissues. In the process, EPCs are exposed to the shear stress generated by blood flow and tissue fluid flow. Our previous study showed that shear stress promotes differentiation of EPCs into mature endothelial cells. In this study, we investigated whether EPCs differentiate into arterial or venous endothelial cells in response to shear stress. When cultured EPCs derived from human peripheral blood were exposed to controlled levels of shear stress in a flow-loading device, the mRNA levels of the arterial endothelial cell markers ephrinB2, Notch1/3, Hey1/2, and activin receptor-like kinase 1 increased, but the mRNA levels of the venous endothelial cell markers EphB4 and neuropilin-2 decreased. Both the ephrinB2 increase and the EphB4 decrease were shear stress dependent rather than shear rate dependent. EphrinB2 protein was increased in shear-stressed EPCs, and the increase in ephrinB2 expression was due to activated transcription and not mRNA stabilization. Deletion analysis of the ephrinB2 promoter indicated that the cis-element (shear stress response element) is present within 106 bp 5′ upstream from the transcription initiation site. This region contains the Sp1 consensus sequence, and a mutation in its sequence decreased the basal level of transcription and abolished shear stress-induced ephrinB2 transcription. Electrophoretic mobility shift assays and chromatin immunoprecipitation assays showed that shear stress markedly increased binding of Sp1 to its consensus sequence. These results indicate that shear stress induces differentiation of EPCs into arterial endothelial cells by increasing ephrinB2 expression in EPCs through Sp1 activation.


Author(s):  
T. Bulduk ◽  
A. U. Yalcin ◽  
O. M. Akay ◽  
S. G. Ozkurt ◽  
H. U. Teke ◽  
...  

Anemia is a common complication of chronic kidney disease (CKD). The most common cause of anemia in CKD is erythropoietin deficiency; and the most important cause of mortality in CKD patients is atherosclerotic vascular complications which are associated with endothelial damage. One of the methods evaluating vascular integrity is the cytometric measurement of circulating endothelial cells and endothelial progenitor cells in peripheral blood. The study aimed to investigate the effects of erythropoietin therapy on endothelial dysfunction by evaluating circulating endothelial cells and endothelial progenitor cells in peripheral blood using the technique of flow cytometry. Methods. A total of 55 hemodialysis patients were evaluated in three groups; those having erythropoietin therapy for at least last 3 months (n = 20) / not having erythropoietin for at least the last 3 months (n = 20) and the patients who started erythropoietin treatment during the study (n = 5). The control group consisted of 20 people. Blood values of the 3rd Group were investigated three times as baseline, 2nd week and 8th week CD34 +, CD105 + cells were evaluated as activated circulating endothelial cells; CD133 +, CD146 + cells were evaluated as activated endothelial progenitor cells. Results. There was no difference between the patients and healthy individuals in terms of circulating endothelial cells and endothelial progenitor cells. In the third group, no differences were observed in circulating endothelial cells / endothelial progenitor cell levels at baseline / 2nd and 8th weeks. There was no correlation between erythropoietin and circulating endothelial cells / endothelial progenitor cells. Conclusion. A correlation is not available between the therapeutic doses of erythropoietin used in hemodialysis patients and circulating endothelial cells / endothelial progenitor cell levels; supratherapeutic doses could change the results.


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.


Circulation ◽  
2007 ◽  
Vol 116 (suppl_16) ◽  
Author(s):  
Nana-Maria Heida ◽  
Marco R Schroeter ◽  
I-Fen Cheng ◽  
Elena I Deryugina ◽  
Thomas Korff ◽  
...  

Endothelial progenitor cells (EPC) have been reported to contribute to neovascularization. We have previously shown that the adipocytokine leptin may enhance the adhesive properties of EPC by upregulating specific integrins. To investigate whether the angiogenic effects of leptin may be mediated by modulation of EPC function, mononuclear cells were isolated from healthy human volunteers and cultivated under endothelial cell conditions for 7 days. In the matrigel assay, pretreatment of EPC with recombinant leptin for 24 hours dose-dependently enhanced their incorporation into tubular structures provided by mature endothelial cells. For example, 138.3 ± 7.6% (P = 0.001) and 145.3 ± 5.5% (P = 0.0001) CM-DiI-labeled EPC were detected after stimulation with 10 and 100 ng/mL leptin, respectively (control-treated EPC defined as 100%). Furthermore, in the spheroid angiogenesis assay, stimulation of EPC with 10 ng/mL leptin increased the number of sprouts (P < 0.0001) and tube length (P < 0.0001) of coincubated mature endothelial cells, and the outgrowth of EPC (P < 0.0001). Addition of 100-fold excess of leptin-neutralizing or leptin-receptor-binding antibodies completely reversed these effects. Moreover, EPC adhesion onto endothelial cell tubules could be reduced by addition of RGD peptides (from 159 ± 13.7% to 101.8 ± 14.6%; P = 0.02), or of neutralizing antibodies against αvβ3 (from 165.3 ± 11.8% to 103.8 ± 13.3%; P = 0.006) or αvβ5 (to 93.5 ± 15.8%; P = 0.005). Further experiments using specific signal transduction inhibitors (10 μM of LY294002, PD98059, or SB203580), as well as Western blot analysis, revealed that leptin signaling in EPC involves phosphoinositide-3 kinase and p42/44, but not by p38 MAP kinase. The effects of leptin could also be confirmed under in vivo conditions. Stimulation of EPC with 100 ng/mL leptin potentiated the insprout of newly formed avian vessels into collagen onplants placed on the chorion allantoic membrane of chicken embryos (angiogenic index, 0.58 ± 0.24) compared to control-treated EPC (0.44 ± 0.27; P = 0.07) and endothelial basal medium alone (0.31 ± 0.26; P = 0.0007). Thus, our in vitro and in vivo results suggest that the angiogenic effects of leptin may partly depend on its specific interaction with endothelial progenitor cells.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1474-1474 ◽  
Author(s):  
Michail Zaboikin ◽  
Tatiana Zaboikina ◽  
Carl E. Freter ◽  
Narasimhachar Srinivasakumar

Abstract Gene and cell therapy for hemophilia A requires the use of the appropriate target cell for genetic modification and, given the advances in genome editing, an approach that can be applied universally for the wide variety of genetic mutations in the coagulation factor VIII gene (F8) responsible for hemophilia A. Recent studies using two different conditional knockout mouse models showed that the principal, and possibly exclusive, source for FVIII in the circulation are endothelial cells (Everett LA et al. Blood 2014.123: 3697; Fahs SA et al. Blood 2014.123: 3706). Since endothelial cells are present in all the major organs previously thought to produce coagulation factor VIII (FVIII), these studies provide the basis for the earlier reports indicating different tissues (liver, lung, spleen, lymphatic tissue) as sources of FVIII. ECs conveniently express von Willebrand factor (vWF) that is essential for the stability of FVIII. The precursor of ECs, endothelial progenitor cells (EPCs), have been isolated from adult human peripheral blood and cord blood. EPCs can readily integrate into existing vascular system upon intravenous injection. EPCs are quite rare in peripheral blood (about 20 colony forming cells per 100 mL of blood). Moreover, EPCs derived from adult peripheral blood have lower proliferative potential than those obtained from cord blood (Ingram DA. Blood. 2004. 104: 2752). In contrast, induced pluripotent stem cells (iPSCs), that are more amenable for expansion, can be readily differentiated into EPCs. Studies have also shown that iPSC-derived EPCs when injected intrahepatically in mice integrated into liver sinusoids, resulted in therapeutic levels of FVIII production (Xu D et al. PNAS 2009. 106: 808). Here we describe an optimized in vitro differentiation protocol for derivation of EPCs from iPSCs. We have previously reported the generation and characterization of human iPSCs from lung fibroblasts (Srinivasakumar et al. PeerJ. 2013;1:e224). In this study we used human iPSCs generated from adult dermal fibroblasts using Yamanaka's non-integrating Epstein-Barr based episomal vectors. We used a step-wise differentiation protocol for obtaining EPCs that was a combination of a method for differentiation of iPSCs into hematopoietic progenitors (Fig A, Steps 1 & 2) to generate hemangioblasts (Niwa A et al. PLoS ONE 6(7): e22261), and a protocol for obtaining EPCs from peripheral blood (Step 3) (Mead LE et al. Current Protocols in Stem Cell Biol. 2C.1.1-2C.1.27). A sorting step after differentiation into hemangioblasts followed by a final round of sorting after step 3 yielded >90% pure population of EPC that exhibited the canonical cell surface markers: CD31 and CD34, and absence of CD45 (Fig B). The cells also took up fluorophore-conjugated acetylated LDL (acLDL-A488) that was inhibited with 50x excess of unlabeled acLDL (Fig C). Immunofluorescence staining for vWF revealed characteristic staining reminiscent of Weibel-Palade bodies in the cytoplasm (Fig D). The cells exhibited the typical tube formation ability in Matrigel (Fig E). Additional studies are needed to determine the proliferative potential of these cells and their ability to integrate into vasculature. To address the myriad mutations shown to be responsible for hemophilia A, we have designed high efficiency dimeric guide RNAs (as part of a separate study) (Zaboikin M et al. Manuscript in preparation) for use with the CRISPR/dCas9-Fok1 system (Tsai SQ et al. Nat Biotechnol. 2014. 32:569) for precise modification at the F8 locus downstream of the first coding exon. We also showed in that study the replacement of target sequence at the site with that of a donor template sequence with desired attributes. We hypothesize that using a donor template that encodes the F8 promoter driving a functional F8 cDNA for homology directed repair at the target double stranded break site will provide an universal solution for the large variety of mutations observed in hemophilia A. Results of genome editing of iPSCs using the above mentioned CRISPR/dCas9-Fok1 system (together with the donor template) followed by the differentiation of genetically modified iPSCs into EPCs will be presented. Figure Figure. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (5) ◽  
pp. 1525-1531 ◽  
Author(s):  
David A. Ingram ◽  
Noel M. Caplice ◽  
Mervin C. Yoder

Abstract The field of vascular biology has been stimulated by the concept that circulating endothelial progenitor cells (EPCs) may play a role in neoangiogenesis (postnatal vasculogenesis). One problem for the field has been the difficulty in accurately defining an EPC. Likewise, circulating endothelial cells (CECs) are not well defined. The lack of a detailed understanding of the proliferative potential of EPCs and CECs has contributed to the controversy in identifying these cells and understanding their biology in vitro or in vivo. A novel paradigm using proliferative potential as one defining aspect of EPC biology suggests that a hierarchy of EPCs exists in human blood and blood vessels. The potential implications of this view in relation to current EPC definitions are discussed.


Sign in / Sign up

Export Citation Format

Share Document