Hypoxia-induced proliferation of tissue-resident endothelial progenitor cells in the lung

2015 ◽  
Vol 308 (8) ◽  
pp. L746-L758 ◽  
Author(s):  
Rintaro Nishimura ◽  
Tetsu Nishiwaki ◽  
Takeshi Kawasaki ◽  
Ayumi Sekine ◽  
Rika Suda ◽  
...  

Exposure to hypoxia induces changes in the structure and functional phenotypes of the cells composing the pulmonary vascular wall from larger to most peripheral vessels. Endothelial progenitor cells (EPCs) may be involved in vascular endothelial repair. Resident EPCs with a high proliferative potential are found in the pulmonary microcirculation. However, their potential location, identification, and functional role have not been clearly established. We investigated whether resident EPCs or bone marrow (BM)-derived EPCs play a major role in hypoxic response of pulmonary vascular endothelial cells (PVECs). Mice were exposed to hypoxia. The number of PVECs transiently decreased followed by an increase in hypoxic animals. Under hypoxic conditions for 1 wk, prominent bromodeoxyuridine incorporation was detected in PVECs. Some Ki67-positive cells were detected among PVECs after 1 wk under hypoxic conditions, especially in the capillaries. To clarify the origin of proliferating endothelial cells, we used BM chimeric mice expressing green fluorescent protein (GFP). The percentage of GFP-positive PVECs was low and constant during hypoxia in BM-transplanted mice, suggesting little engraftment of BM-derived cells in lungs under hypoxia. Proliferating PVECs in hypoxic animals showed increased expression of CD34, suggesting hypoxia-induced gene expression and cell surface antigen of EPC or stem/progenitor cells markers. Isolated PVECs from hypoxic mice showed colony- and tube-forming capacity. The present study indicated that hypoxia could induce proliferation of PVECs, and the origin of these cells might be tissue-resident EPCs.

2009 ◽  
Vol 50 (5) ◽  
pp. 469-475 ◽  
Author(s):  
Nuttawut SERMSATHANASAWADI ◽  
Hideto ISHII ◽  
Kaori IGARASHI ◽  
Masahiko MIURA ◽  
Masayuki YOSHIDA ◽  
...  

2013 ◽  
Vol 13 (02) ◽  
pp. 1350050 ◽  
Author(s):  
ZHI-QIANG YAN ◽  
YU-QING LI ◽  
BIN-BIN CHENG ◽  
QING-PING YAO ◽  
LI-ZHI GAO ◽  
...  

Differentiation of endothelial progenitor cells (EPCs) plays important roles in endothelial repair after vessel injury. Endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and mechanical forces, including cyclic strain and shear stress, synergistically form the microenvironment of EPCs. However, the synergistic effect of cyclic strain, ECs, and VSMCs on the differentiation of EPCs remains unclear. In the present study, EPCs were indirectly co-cultured with stretched ECs or VSMCs that were subjected to 5%, 1.25-Hz cyclic strain by using FX-4000T Strain Unit. Then, Western blot and real-time PCR were used to examine expressions of EC marker, i.e., vascular cell adhesion molecule (VCAM), CD31, von Willebrand factor (vWF); VSMC markers, i.e., α-actin, Calponin, and SM22α; and signaling molecules, i.e., p-Akt and p-ERK. In static, co-cultured ECs increased expression of VCAM and phosphorylation of Akt and ERK in EPCs compared to that in EPCs cultured alone. In EPCs, co-cultured VSMCs decreased expressions of CD31 and vWF, but increased expressions of Calponin and SM22α. Stretched ECs reduced expressions of CD31 and vWF, enhanced Calponin and SM22α, and repressed phosphorylations of Akt and ERK in EPCs. Stretched VSMCs decreased CD31, increased Calponin and SM22α expressions, and repressed phosphorylation of Akt and ERK in EPCs. Our results suggest that ECs promoted EPC differentiation into ECs in static. VSMCs in static, as well as stretched ECs and stretched VSMCs, promoted EPC differentiation into VSMCs. Phosphorylation of Akt and ERK might be involved in EPC differentiation, mediated by the stretched ECs and VSMCs.


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 ◽  
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.


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.


Circulation ◽  
2014 ◽  
Vol 130 (suppl_2) ◽  
Author(s):  
Yuichi Nakamura ◽  
Satoshi Suzuki ◽  
Takeshi Shimizu ◽  
Makiko Miyata ◽  
Tetsuro Shishido ◽  
...  

Background: High mobility group box 1 (HMGB1) is a DNA-binding protein secreted into extracellular space from necrotic cells and acts as a cytokine. We have reported that HMGB1 attenuates cardiac damage and restores cardiac function by enhancing angiogenesis after myocardial infarction (MI). We examined the role of HMGB1 in angiogenesis from bone marrow (BM) -derived cells in the heart, using transgenic mice with cardiac-specific overexpression of HMGB1 (HMGB1-TG). Methods and Results: HMGB1-TG mice and wild-type littermate (WT) mice were lethally irradiated and injected with BM cells from green fluorescent protein (GFP) mice through the tail vein. Two weeks after BM transplantation, the left anterior descending artery was ligated to create MI. In flow cytometry analysis, GFP-positive cells were identified as donor BM cells-derived endothelial progenitor cells (EPC) if they were positive for both CD34 and CD144 in granulocyte differentiation antigen-1-negative fraction. Circulating EPC mobilized from BM was increased at 1 week after MI in HMGB1-TG mice compared with WT mice (41.9% vs. 24.5%, P < 0.01). Histological examination showed the size of MI was smaller in HMGB1-TG mice than in WT mice (42.9% vs. 59.1%, P < 0.01) at 4 weeks after MI. In myocardial immunofluorescence staining, GFP and CD31 double-positive cells were BM-derived cells engrafted within myocardial tissue as vascular endothelial cells of new capillaries or arterioles. The ratio of these double positive cells to all cardiac cells was significantly higher in the HMGB1-TG mice than in the WT mice (8.3% vs. 2.9%, P < 0.01). Enzyme-linked immunosorbent assay revealed that the levels of cardiac vascular endothelial growth factor at 1 week after MI were higher in HMGB1-TG mice than in WT mice (642.1 vs. 390.7 pg/dl, P < 0.05). Conclusions: The present study demonstrated the direct in vivo evidence that HMGB1 promoted angiogenesis and reduced MI size by enhancing mobilization and differentiation of BM cells to EPC, migration to the border zone of MI, and engraftment as vascular endothelial cells of new capillaries or arterioles in the infarcted heart.


Sign in / Sign up

Export Citation Format

Share Document