Endothelial Cells Derived from Human Vessel Walls Contain a Complete Hierarchy of Endothelial Progenitor Cells.

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.

Blood ◽  
2005 ◽  
Vol 105 (7) ◽  
pp. 2783-2786 ◽  
Author(s):  
David A. Ingram ◽  
Laura E. Mead ◽  
Daniel B. Moore ◽  
Wayne Woodard ◽  
Amy Fenoglio ◽  
...  

AbstractEndothelial progenitor cells (EPCs) can be isolated from adult peripheral and umbilical cord blood and expanded exponentially ex vivo. In contrast, human umbilical vein endothelial cells (HUVECs) or human aortic endothelial cells (HAECs) derived from vessel walls are widely considered to be differentiated, mature endothelial cells (ECs). However, similar to adult- and cord blood–derived EPCs, HUVECs and HAECs derived from vessel walls can be passaged for at least 40 population doublings in vitro. Based on this paradox, we tested whether EPCs reside in HUVECs or HAECs utilizing a novel single cell deposition assay that discriminates EPCs based on their proliferative and clonogenic potential. We demonstrate that a complete hierarchy of EPCs can be identified in HUVECs and HAECs derived from vessel walls and discriminated by their clonogenic and proliferative potential. This study provides evidence that a diversity of EPCs exists in human vessels and provides a conceptual framework for determining both the origin and function of EPCs in maintaining vessel integrity.


2001 ◽  
Vol 42 (4) ◽  
pp. 699-708 ◽  
Author(s):  
Giorgio Lambertenghi Deliliers ◽  
Lorenza Caneva ◽  
Rossella Fumiatti ◽  
Federica Servida ◽  
Paolo Rebulla ◽  
...  

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. 3911-3911
Author(s):  
David A. Ingram ◽  
Amy Fenoglio ◽  
Laura E. Mead ◽  
Mervin C. Yoder

Abstract Human EPCs are currently defined as a CD133+/Flk1+ subpopulation of CD34+ cells that can be isolated from peripheral blood, bone marrow, and fetal liver (Nature Medicine, 2003). While these cells are widely considered to be EPCs and subsequently used for angiogenic therapies and biomarkers of cardiovascular disease, their proliferative and clonogenic potential has not been compared to other populations of EPCs, which do not express the cell surface antigens, CD34 and AC133. Analogous to a paradigm established in the hematopoietic cell system, we recently developed a single cell clonogenic 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 (Blood, 2004). Utilizing this assay and EPCs derived from human umbilical cord blood mononuclear cells, we compared the clonogenic and proliferative potential of 1,000 single CD34+AC133+Flk1+ and CD34-AC133−Flk1+ umbilical cord blood derived endothelial cells. We conducted four independent experiments. We demonstrate that a complete hierarchy of EPCs can be identified in a population of CD34−AC133−Flk1+ cord blood derived ECs (Table I, n=4). Remarkably, we further show that CD34−AC133−Flk1+ cord blood derived ECs contain more proliferative EPCs (LPP-ECFCs and HPP-ECFCs) compared to CD134+AC133+Flk1+ cord blood derived ECs (Table I, n=4, *p<0.05). In fact, preliminary experiments suggest that CD34−AC133−Flk1+ cells mature into CD34+AC133+Flk1+ cells. Finally, some individual single CD34−AC133−Flk1+ HPP-ECFCs can expand rapidly to 107 cells in ex vivo culture. Retroviral marking of the cell progeny derived from the single cell confirmed that they were derived from the parent cell. Thus, these studies describe the use of a clonogenic assay to identify a novel population of CD34−AC133−Flk1+ EPCs, and preliminary experiments demonstrate that CD34 and AC133 are not universal cell surface markers of the most primitive EPCs. Percent of 1,000 Single Cells Plated Mature EC EC-Cluster LPP-ECFC HPP-ECFC CD34+AC133+Flk1+ ECs 70±11 16±5 10±4 3±1 CD34-AC133-Flk1+ ECs 52±12 17±6 20±3* 10±4*


2019 ◽  
Vol 14 (6) ◽  
pp. 460-465 ◽  
Author(s):  
Jing Jia ◽  
Baitao Ma ◽  
Shaoshuai Wang ◽  
Ling Feng

Endothelial progenitor cells (EPCs) are implicated in multiple biologic processes such as vascular homeostasis, neovascularization and tissue regeneration, and tumor angiogenesis. A subtype of EPCs is referred to as endothelial colony-forming cells (ECFCs), which display robust clonal proliferative potential and can form durable and functional blood vessels in animal models. In this review, we provide a brief overview of EPCs’ characteristics, classification and origins, a summary of the progress in preclinical studies with regard to the therapeutic potential of human umbilical cord blood derived ECFCs (CB-ECFCs) for ischemia repair, tissue engineering and tumor, and highlight the necessity to select high proliferative CB-ECFCs and to optimize their recovery and expansion conditions.


2007 ◽  
Vol 29 (6) ◽  
pp. 388-392 ◽  
Author(s):  
Javier Garc??a-Castro ◽  
Antonio Balas ◽  
Manuel Ram??rez ◽  
Antonio P??rez-Mart??nez ◽  
Luis Madero ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (10) ◽  
pp. 3360-3362 ◽  
Author(s):  
Karen M. Pastos ◽  
William B. Slayton ◽  
Lisa M. Rimsza ◽  
Linda Young ◽  
Martha C. Sola-Visner

Abstract Umbilical cord blood (CB) is a valuable source of stem cells for transplantation, but CB transplantations are frequently complicated by delayed platelet engraftment. The reasons underlying this are unclear. We hypothesized that CB- and peripheral-blood (PB)–derived megakaryocytes (MKs) respond differently to the adult hematopoietic microenvironment and to thrombopoietin (Tpo). To test this, we cultured CB- and PB-CD34+ cells in adult bone marrow stromal conditioned media (CM) or unconditioned media (UCM) with increasing concentrations of recombinant Tpo and compared the effects of these conditions on CB-versus PB-MKs. PB-MKs reached highest ploidy in response to UCM + 100 ng/mL rTpo, and the addition of CM inhibited their maturation. In contrast, CB-MKs reached highest ploidy in CM without rTpo, and high rTpo concentrations (> 0.1 ng/mL) inhibited their maturation. This is the first evidence that human neonatal and adult MKs have substantially different biologic responses to Tpo and potentially to other cytokines.


Sign in / Sign up

Export Citation Format

Share Document