Contribution of endothelial progenitor cells to neovascularization (Review)

  • Authors:
    • Da-Wei Li
    • Zhi-Qiang Liu
    • Jun Wei
    • Ying Liu
    • Lin-Sen Hu
  • View Affiliations

  • Published online on: August 23, 2012     https://doi.org/10.3892/ijmm.2012.1108
  • Pages: 1000-1006
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endothelial progenitor cells (EPCs) are a cell population mobilized from bone marrow into the peripheral circulation and recruited into sites of vessel injury to participate in blood vessel formation in both physiological and pathological conditions. Due to the lack of unique surface markers and different isolation methods, EPCs represent heterogeneous cell populations including cells of myeloid or endothelial origin. Evidence suggests that EPCs play a critical role in postnatal blood vessel formation and vascular homeostasis and provide a promising therapy for vascular disease. However, the mechanisms by which EPCs participate in new vessel formation are still incompletely understood. We review the process of EPCs in neovascularization including EPC mobilization, migration, adhesion and effect on new vessel formation, in an attempt to better understand the underlying mechanisms and to provide potential effective management for the treatment of patients with vascular disease.

Contents

Introduction

Putative endothelial progenitor cells

Mobilization of endothelial progenitor cells

Migration of endothelial progenitor cells

Endothelial progenitor cell adhesion

Endothelial progenitor cells in new blood vessel formation

Introduction

Vascular disorders, possessing high morbidity and mortality, are suffered by a multitude of patients due to the limitations of current therapies. Efficient repair of damaged endothelium and enhancement of the ability to form new blood vessels are crucial for treatment of these disease. Disruption of endothelial integrity initiates proliferation and migration of endothelial cells (ECs) from adjacent preexisting blood vessels, which promotes reendothelialization and neointimal formation of vascular lesions. The postnatal formation of new vessels occurs exclusively through proliferation and migration of existing neighboring ECs, referred to as angiogenesis. This traditional concept of blood vessel formation has been challenged by Asahara et al (1), who first described the cell population derived from bone marrow contributing to new vessel formation and termed these cells endothelial progenitor cells (EPCs). In contrast to angiogenesis, the formation of new blood vessels via mature ECs from proliferation and differentiation of bone marrow-derived progenitor cells is defined as vasculogenesis. Therefore, EPCs proposed as a promising treatment for vascular disease have been extensively studied by many investigators. However, due to the absence of specific surface markers, these progenitor cells represent heterogeneous cell populations including cells of myeloid or endothelial origin (2,3). Recently, three types of cells have been most studied as EPCs including colony-forming unit-Hill (CFU-Hill) cells, circulating angiogenic cells (CACs) and endothelial colony-forming cells (ECFCs). CFU-Hill cells and CACs are usually referred to as early outgrowth EPCs according to their obtention from short term blood sample culture. In contrast, ECFCs are termed as late outgrowth EPCs. Although the contribution of endothelial progenitor cells including early outgrowth EPCs and late outgrowth EPCs to new vessel formation has been established, the underlying mechanisms of bone marrow EPC-induced vascularization remain unclear, and need to be further studied.

Putative endothelial progenitor cells

Putative EPCs encompass different cell populations mainly containing hematopoietic and endothelial progenitor cells. These different populations result in a mixed ability to enhance the formation of new blood vessels (2,4). Recently, there are mainly three culture methods used to isolate EPCs.

The first method originally described by Asahara et al (1) has been modified (5,6), and can be performed using a commercially available kit. Mononuclear cells (MNCs) isolated from adult peripheral blood (PB) or cord blood (CB) are plated on fibronectin-coated tissue culture surfaces. After 48 h, the adherent macrophages and mature ECs in the sample are depleted, and the nonadherent colony cells are replated on fresh fibronectin-coated dishes. Over the next 5–9 days, adherent colonies emerge centrally, comprised of round cells with spindle-shaped cells sprouting at the periphery. These colonies are often referred to as colony-forming unit-Hill (CFU-Hill) or CFU-ECs. CFU-Hill cells have been shown to express the cell surface antigens CD31, CD105, CD144, CD146, vWF and KDR (VEGFR2). These cells also display the ability to ingest acetylated low-density lipoprotein and binding of the lectin Ulex europeus agglutinin-1 (UEA-1). These phonotypes and functions are often ascribed to ECs. CFU-Hill cells also express several monocyte/macrophage markers including CD14 and CD115 and hematopoietic-specific cell surface antigen CD45, and display non-specific esterase activity (13,7). Recently, evidence indicates that these progenitor cells originate from hematopoietic origins (3,8), and enhance new blood vessel formation by paracrine action (4,9).

Another widely used method to isolate putative EPCs involves the culture of PB MNCs in supplemented endothelial growth media for 4 days, and the nonadherent cells are washed away, thus an adherent cell target population remains (7,10). These adherent cells, referred to as CACs, have been shown to express the endothelial cell surface antigens, CD31, CD144, vWF and KDR, bind Ulex Europaeus selectin (7,10,11), and have been shown to uptake acetylated low-density lipoprotein (ac-LDL) (10). This particular method of CAC isolation and culture is contaminated by platelets, and these platelet membrane proteins may be transferred to the target cells by adhering to MNCs in the culture. CFU-Hill cells and CACs are also referred to as early outgrowth EPCs.

The third method of culture yields isolation and identification of ECFCs. In this method, adult PB MNCs or umbilical cord blood (UCB)-derived MNCs are collected and plated onto collagen I-coated plates in endothelial-specific growth media (3,12). Non-adherent cells are discarded during gentle washing steps. ECFC colonies emerge from the adherent cell population 10–21 days for PB and 5–7 for UCB after plating and display a cobblestone EC appearance (3,1214). Some studies have demonstrated that ECFCs express the cell surface antigens, CD31, CD105, CD144, CD146, vWF and KDR and uptake ac-LDL (3,12). In contrast to CFU-Hill cells, ECFCs do not express the hematopoietic or monocyte/macrophage cell surface antigens, CD14, CD45 or CD115 (3), and exhibit robust capacity in proliferation and forming secondary endothelial cell colonies. Further studies showed that ECFCs possess the ability of forming capillary-like in vitro and perfused blood vessels in vivo (3,15).

The process of EPCs in the formation of new blood vessels include multiple steps in which many cytokines and modulators are involved. Although there has been a great deal of progress in this field, a comprehensive understanding of mechanisms by which EPCs participate in neovascularization will contribute to the development of effective therapies for vascular disorders.

Mobilization of endothelial progenitor cells

It has been indicated that levels EPCs in peripheral circulation are low under physiological conditions, and that these cells may reside in the bone marrow niche. The majority of EPCs are quiescent and tethered by integrins to stromal cells in a microenvironment within the bone marrow (16,17). The mobilization of EPCs from the bone marrow into the peripheral circulation is the crucial step for these cells to participate in postnatal vasculogenesis. The precise mechanism of EPC mobilization is not entirely elucidated and is still under investigation. It has been demonstrated that these cells can be converted into functional cells and released from the stem cell niche in response to various special cytokines and factors.

Vascular endothelial growth factor (VEGF), a pleiotropic cytokine, functions in neovascularization as an endothelial cell mitogen (18), chemotactic agent (19) and inducer of vascular permeability (2022). Further studies have demonstrated that VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived EPCs (23), and VEGF gene transfer has been shown to augment circulating EPCs in human subjects (24). In burn injures, VEGF levels are elevated in the plasma and are responsible for the enhancement of mobilization of VEGFR2+ EPCs in circulation (25). In addition, VEGF has the ability to upregulate the levels of granulocyte colony-stimulating factor (G-CSF) (26), which can induce the release of progenitor cells from bone marrow (27). The interaction between VEGF and VEGFR leads to the activation of bone marrow NOS, then produces nitric oxide (NO), which is responsible for MMP-9 activation. The activated MMP-9 contributes to the release of soluble kit ligand (sKitL) which enhances the mobility of VEGFR2+ EPCs and stimulates the mobilization of these cells from bone marrow to the peripheral circulation (28).

The CXC chemokines, playing a role as significant regulators, have been implicated in the mobilization of bone marrow-derived stem or progenitor cells. Stromal cell-derived factor-1 (SDF-1) is the most characterized player in EPC mobilization and a potent chemokine in EPC adhesion and migration, therefore it exerts a beneficial effect on neovascularization. A range of stimuli such as inflammation and hypoxia account for upregulated levels of SDF-1 expressed in the extra-cellular matrix (29). Ischemic environments appear to promote the release of SDF-1, in proportion to the degree of hypoxia, with elevated intracellular SDF-1 mRNA expression in ischemic ECs (29). This occurs within the first hour of ischemia (30). The elevation of SDF-1 in plasma stimulates mobilization of CXCR4+ bone marrow cells, including hematopoietic stem cells (HSCs) and EPCs (28,31). CXCR4 is a receptor of SDF-1 highly expressed by hematopoietic progenitor and endothelial progenitor cells (32,33). The interaction of SDF-1 and CXCR4 not only initiates the mobilization of EPCs from bone marrow, but stimulates the recruitment and retention of stem cells to ischemic areas (17,34,35). SDF-1 can be secreted by platelets and induce chemotaxis of EPCs (36). EPCs themselves can also release SDF-1 in a paracrine fashion (37). The first response to vascular injury is the adhesion of platelets to the exposed subendothelium (3841), which provides the signals of target for the mobilization and homing of stem cells to the damaged area (42). SDF-1α gene transfer promotes the mobilization of EPCs into the peripheral blood and enhances neovascularization in ischemic animal models. However, the effect of SDF-1 on the mobilization of EPCs is ablated in the absence of injury. Furthermore, blockade of VEGF or NOS signaling prevents all SDF-1-induced effects, indicating that VEGF/eNOS signaling is involved in the benefit from SDF-1 upregulation in neovascularization (43). Studies have shown that the mobilization of bone marrow-derived stem or progenitor cells requires the activation of bone marrow NOS, which are responsible for releasing progenitor cells from bone marrow via the NO-MMP-9-soluble kit ligand cascade (28,44). SDF-1 also enhances the mobilization of EPCs by upregulating the levels of VEGF which contribute to the release of EPCs from the bone marrow to the peripheral circulation. Interleukin-8 (IL-8) is an inflammatory chemokine originally described as a chemotactic factor for leukocytes. Recent data have implicated IL-8 as a regulater in mobilizing EPCs into the peripheral circulation by binding both CXCR1 and CXCR2. This function is synergized by G-CSF in animal models (45,46).

Nitric oxide (NO) initially discovered as an endothelium-derived relaxing factor plays a key role in regulating the physiological properties of blood vessels, including vasodilation, vascular permeability, and antithrombotic properties (47). NO also participates in maintaining vascular integrity and blood flow by modulating platelet-endothelial interactions (48). In addition to vasoprotective effects, NO is now recognized as a key determinant in EPC mobilization from the bone marrow into circulation resulting in enhancement of ischemic limb perfusion and wound healing (49,50). Production of NO relies on the conversion of L-arginine to L-citrulline catalyzed by nitric oxide synthases (51,52). The enzyme has four isoforms, NOS1 or neuronal nitric oxide synthase (nNOS), NOS2 or inducible nitric oxide synthase (iNOS), NOS3 or endothelial nitric oxide synthase (eNOS) and NOS4 or mitochondrial nitric oxide synthase (mtNOS). mtNOS recently identified appears to be a constitutively active eNOS-like isoform (53). Among these, eNOS selectively expressed in vascular endothelial cells and surrounding stromal cells plays a central role in vascular biology. NO has also been shown to be expressed by various EPC subtypes. eNOS plays a crucial role in regulation of mobilization and function of EPCs (44). Many patients with diabetes suffer from delayed or nonhealing wounds of the lower extremities and diabetic foot ulcers. Likely, EPCs which play a crucial role in postnatal neovascularization are impaired by hyperglycemia in diabetes. Hyperglycemia and diabetes are associated with impaired eNOS function which results in depressed EPC mobilization into circulation. The decreased levels of phosphorylated eNOS, but not the level of eNOS protein, was found to be responsible for impaired mobilization of EPCs (54). It is known postnatally that stem cells including EPCs reside in the bone marrow via adherence to stromal cells in the stem cell niche by integrins and can be released into the peripheral circulation in response to cytokines and other angiogenic factors (16,17). The mechanisms of EPC mobilization from bone marrow are still incompletely understood. The NO-MMP-9-sKitL-ckit cascade may play a central role in this process. eNOS can be stimulated in bone marrow by many cytokines involved in the event, then NO is produced. NO can stimulate MMP-9, which result in the release of sKitL from the stromal cell membrane-bound kit ligand (mKitL). c-Kit expressed by EPCs contributes to the retention of EPCs within the bone marrow niche. c-Kit is also the receptor for sKitL and can be released from bone marrow in response to binding to sKitL, resulting in mobilization of c-Kit+ EPCs from the cell niche into circulation (Fig. 1). Certainly, the mechanisms associated with the mobilization of EPCs induced by these factors in the process of new blood vessel formation required further study.

Migration of endothelial progenitor cells

EPCs are released from bone marrow and recruited to sites of injury to enhance new blood vessel formation and wound healing. However, the signals that target EPCs to the sites of vascular injury are still poorly understood. Chemokines are important factors controlling cellular migration. The CXC chemokines have been involved in the field of vascularization because of their ability to modulate the functions of vascular endothelial cells and EPCs by increasing the migratory capacity of these cells to the sites of injured vessels. SDF-1 is the most potent chemoattractant of EPCs. The formation of SDF-1 concentration gradients from the periphery to ischemia plays a pivotal role in the migration of EPCs (Fig. 2). CXC receptor (CXCR4) is the predominant receptor for SDF-1 and is selectively expressed in vascular endothelial cells and EPCs (29,55). Studies have demonstrated that SDF-1 is able to track various types of CXCR4+ cells (29,56,57). It is upregulated in ischemic tissue and acts as a homing signal for EPCs (58). The migration of EPCs is significantly reduced by anti-CXCR4 neutralizing antibodies (29). Further evidence suggest that SDF-1-induced EPC migration is mediated through the PI3K/Akt/eNOS signal transduction pathway (59). IL-6 is a multifunctional cytokine which possesses the ability to modulate cell proliferation and differentiation in physiological conditions (6062). Studies indicate that IL-6 contributes to cerebral EC and smooth muscle cell (SMC) proliferation and migration in vitro (6365). Furthermore, IL-6 in vitro was found to enhance EPC proliferation and migration in a dose-dependent manner. EPCs express IL-6 receptor (gp80 and gp130), and the molecular mechanism of EPC proliferation and migration is mediated by IL-6 via gp80/gp130 signaling pathways including downstream ERK1/2 and STAT-3 phosphorylation (66). The signal pathways widely exist in many cell types including EPCs. Activation of the ERK1/2 and STAT-3 pathways plays a crucial role in EC proliferation, migration and microvascular tube formation (65,67,68). This process may include the modulation of cytoskeletal reorganization in EPCs, which is associated with cell migration. Studies also demonstrate that activation of the CXCR1 is transient and characterized by stress fiber formation, whereas activation of CXCR2 is prolonged and associated with cell contraction (69). Additionally, pleiotrophin (PNT), produced under ischemic conditions, also chemotactically attracts early EPCs to sites of vessel injury (70).

It is well known that cell mobility is closely associated with the cytoskeleton. Various cell activities, including migration, morphological change, and polarity formation are regulated by actin filament dynamics including actin filament disassembly and severing and reorganization. During cell migration, actin filaments are assembled to form the lamellipodial protrusion at the leading edge of the cell. Addition of actin monomers to the barbed ends of actin flaments at the tip of the lamellipodium provides the force with which to propel the membrane forward. These events are modulated by a variety of actin-binding proteins (71,72). As an important actin-binding protein, cofilin modulates actin filament assembly and disassembly, especially in stimulus-induced lamellipodium formation. Our group studied in vitro the mechanism of the impaired migration of EPCs induced by H2O2 using proteomic analysis, and indicated that the oxidative levels of actin and/or cofilin, not its number, are responsible for the attenuated ability of EPC migration (73). We hypothesized that modulators involved in the migration of EPCs activate actin-binding proteins which regulate the actin filament and result in the migration of EPCs into areas of peripheral ischemic or wound tissue. Further studies are needed to more specifically identify the underlying mechanism of EPC migration.

Endothelial progenitor cell adhesion

Accumulating data indicate that CD34+ bone marrow-derived progenitor cells recruit and differentiate into ECs in the vascular intima in response to vessel injury (7477). The mechanisms involved in the recruitment of EPCs from the circulation to adhesion to the sites of neovascularization, although incompletely understood, appear to mainly rely on the interaction between P selectin glycoprotein ligand-1 (PSGL-1) expressed on EPCs and P-selectin expressed on platelets (Fig. 3). Vascular injury initiates the adhesion of platelets to the exposed subendothelium (3841). Upon adhesion, platelets become activated and express P-selectin and release SDF-1, which are both implicated in the adhesion of EPCs to the sites of vascular injury (42). SDF-1 released into the microenvironment of sites of vascular injury may play a crucial role in the modulation of this process via the SDF-1/CXCR4 pathway (58).The interaction of SDF-1 and CXCR4 upregulates the PSGL-1 expression on the surface of EPCs, which is the major ligand of P-selectin. The bonding contributes to the adhesion of EPCs to the sites of vessel injury and enhances their pro-angiogenic capacity. The ability of EPCs homing to the ischemic myocardium is impared by the inhibition of the SDF-1/CXCR4 pathway (78), and the adhesion of EPCs to the sites of injury is also significantly inhibited by CXCR4 blockade as noted in a model of hindlimb ischemia (32). Interestingly, SDF-1 30 min after vascular injury is highly expressed in the aggregated platelets at the sites of vessel foci, whereas 4 h after endothelial disruption, SDF-1 is abundantly expressed in both SMCs and the aggregated platelets (42). It seems that platelets constitute the initial and presumably more short-lived source of SDF-1 that first directs EPCs to the site of vessel damage. In contrast, SMCs appear to account for the long-term SDF-1 release over the ensuing days and weeks after vessel injury required to sustain the process of vascular remodeling and repair (79,80). Integrins, a type of cell adhesion molecule, regulate cell adhesion and migration by interaction with the extracellular matrix. EPCs selectively express β1- and β2-integrins which contribute to the homing of these cells by strengthening adhesion to the damaged endothelial monolayer and by enhancing migration into the endothelial monolayer (81). High-mobility group box 1 (HMGB1) released into the extracellular space by necrotic cells also participates in EPC adhesion to ischemic areas via activation of β1- and β2-integrins (82). ECFCs, however, are not of hematopoietic origin and do not express β2-integrins in contrast to early EPCs; the homing of these cells to the ischemic zone may be related to the expression of E-selectin under specific conditions (83). GPIIb-dependent platelet aggregation plays a critical role in the recruitment of EPCs at sites of vascular injury. GPIIb may promote the homing of EPCs into vascular foci by forming a link between platelets and EPCs using a GPIIb-dependent bridging mechanism previously reported to occur during platelet-EC interactions (84). Furthermore, platelets aggregated by GPIIb can form a cross-linking structure in which PCs get trapped (42). In addition, α4-integrin seems to participate in circulating progenitor cell homing to the sites of neovascularization and improves blood perfusion in impaired tissue (85,86).

Endothelial progenitor cells in new blood vessel formation

The contribution of EPCs to vascularization has been demonstrated in animal models and in humans. Infusion of EPCs, capillary density and new vessel formation are augmented in impaired tissues. In ischemic animal models, injection of EPCs was found to significantly enhance neovascularization in the sites of foci thus improving blood flow and injured tissue recovery (8790). In human trials, transplantation of EPCs generated a trend towards functional improvement in patients with acute myocardial infarction (91,92). These studies indicate that EPCs play a pivotal role in new blood vessel formation. However, putative EPCs and their role in neovascularization remain controversial. These controversies may be due to discrepancies in their identification and the complex mechanisms involved in EPC-induced neovascularization. Putative EPCs encompass different cell populations including hematopoietic or endothelial origin. Accumulating data suggest that hematopoietic cells facilitate new blood vessel formation without direct incorporation into the endothelial intima in the process of postnatal vasculogenesis, and mainly depend on humoral and cell-mediated support functions (9396). In contrast, the contribution of late outgrowth endothelial cells (ECFCs) to neovascularization may combine the incorporation into newly formed vessels and the release of pro-angiogenic factors in a paracrine manner, although there is less direct evidence showing that vascular neointimal formation occurs via mature ECs from proliferation and differentiation of EPCs. Studies have shown that EPCs are essential in wound healing by facilitating new vessel formation (54,97). Delayed wound healing in diabetes results from impaired eNOS activition and hence reduces mobilization of EPCs from the bone marrow into circulation, which can be reversed by the treatment of hyperoxia. However, increased levels of circulating EPCs induced by hyperoxia fail to enhance wound healing. One possible explanation is that the downregulated production of SDF-1 in local wound lesions in diabetes is responsible for the ablated effects. Administration of SDF-1 into wounds improves efficiency of EPC migration consequently enhancing nevascularization and wound healing (54), indicating that thorough understanding of the mechanisms of EPC-mediated new vessel formation are required. These unanswered questions need to be further elucidated.

In conclusion, the contribution of EPCs to neovascularization provides a promising therapeutic target for treating patients with vascular disorders. However, the putative EPCs are heterogeneous cell populations and demonstrate a mixed capacity of contribution to vessel formation. Furthermore, vasculogenesis is a complex process including EPC mobilization from the bone marrow, circulation in blood vessels, recruitment to the impaired sites, adhesion to the injured vessel intimal and exertion effect on new blood formation, which are controlled by multiple cytokines and modulators via different mechanisms, a better understanding of which will elucidate the effect of EPCs in neovascularization and eventually lead to the development of an efficient approach for treating patients with vascular disease.

Abbreviations:

EPCs

endothelial progenitor cells

ECs

endothelial cells

CFU-Hill

colony-forming unit-Hill

CACs

circulating angiogenic cells

ECFCs

endothelial colony-forming cells

MNCs

mononuclear cells

PB

peripheral blood

CB

cord blood

UEA-1

lectin Ulex europeus agglutinin-1

UCB

umbilical cord blood

VEGF

vascular endothelial growth factor

SDF-1

stromal cell-derived factor-1

PNT

pleiotrophin

HMGB1

high-mobility group box 1

SMCs

smooth muscle cells

eNOS

endothelial nitric oxide synthase

References

1. 

T AsaharaT MuroharaA Sulli vanIsolation of putative progenitor endothelial cells for angiogenesisScience275964967199710.1126/science.275.5302.9649020076

2. 

J RehmanJ LiCM OrschellKL MarchPeripheral blood ‘endothelial progenitor cells’ are derived from monocyte/macrophages and secrete angiogenic growth factorsCirculation107116411692003

3. 

MC YoderLE MeadD PraterRedefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principalsBlood10918011809200710.1182/blood-2006-08-04347117053059

4. 

J HurCH YoonHS KimCharacterization of two types of endothelial progenitor cells and their different contributions to neovasculogenesisArterioscler Thromb Vasc Biol24288293200410.1161/01.ATV.0000114236.77009.0614699017

5. 

JM HillG ZalosJPJ HalcoxCirculating endothelial progenitor cells, vascular function, and cardiovascular riskN Engl J Med348593600200310.1056/NEJMoa02228712584367

6. 

H ItoII RoviraML BloomEndothelial progenitor cells as putative targets for angiostatinCancer Res5958755877199910606226

7. 

C KalkaH MasudaT TakahashiTransplantation of ex vivo expanded endothelial progenitor cells for therapeutic neovascularizationProc Natl Acad Sci USA9734223427200010.1073/pnas.97.7.342210725398

8. 

E GunsiliusHC DubaAL PetzerEvidence from a leukaemia model for maintenance of vascular endothelium by bone-marrow-derived endothelial cellsLancet35516881691200010.1016/S0140-6736(00)02241-810905245

9. 

CH YoonJ HurKW ParkSynergistic neovascularization by mixed transplantation of early endothelial progenitor cells and late outgrowth endothelial cells: the role of angiogenic cytokines and matrix metalloproteinasesCirculation11216181627200510.1161/CIRCULATIONAHA.104.503433

10. 

M VasaS FichtlschererA AicherNumber and migratory activity of circulating endothelial progenitor cells inversely correlate with risk factors for coronary artery diseaseCirc Res89E1E7200110.1161/hh1301.09395311440984

11. 

S DimmelerA AicherM VasaHMG-CoA reductase inhibitors (statins) increase endothelial progenitor cells via the PI 3-kinase/Akt pathwayJ Clin Invest108391397200110.1172/JCI20011315211489932

12. 

DA IngramLE MeadH TanakaV MeadeA FenoglioK MortellIdentification of a novel hierarchy of endothelial progenitor cells using human peripheral and umbilical cord bloodBlood10427522760200410.1182/blood-2004-04-139615226175

13. 

Y LinDJ WeisdorfA SoloveyRP HebbelOrigins of circulating endothelial cells and endothelial outgrowth from bloodJ Clin Invest1057177200010.1172/JCI807110619863

14. 

DA IngramLE MeadDB MooreW WoodardA FenoglioMC YoderVessel wall-derived endothelial cells rapidly proliferate because they contain a complete hierarchy of endothelial progenitor cellsBlood10527832786200510.1182/blood-2004-08-3057

15. 

JM Melero-MartinZA KhanA PicardIn vivo vasculogenic potential of human blood-derived endothelial progenitor cellsBlood10947614768200710.1182/blood-2006-12-06247117327403

16. 

T LapidotI PetitCurrent understanding of stem cell mobilization: the roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines, and stromal cellsExp Hematol30973981200210.1016/S0301-472X(02)00883-412225788

17. 

T LapidotA DarO KolletHow do stem cells find their way home?Blood10619011910200510.1182/blood-2005-04-141715890683

18. 

N FerraraWJ HenzelPituitary follicular cells secrete a novel heparin-binding growth factor specific for vascular endothelial cellsBiochem Biophys Res Commun161851858198910.1016/0006-291X(89)92678-8

19. 

A YoshidaB Anand-ApteBR ZetterDifferential endothelial migration and proliferation to basic fibroblast growth factor and vascular endothelial growth factorGrowth Factors135764199610.3109/089771996090345668962720

20. 

A BrkovicMG SiroisVascular permeability induced by VEGF family members in vivo: role of endogenous PAF and NO synthesisJ Cell Biochem100727737200710.1002/jcb.2112417115409

21. 

N FerraraMolecular and biological properties of vascular endothelial growth factorJ Mol Med77527543199910.1007/s00109990001910494799

22. 

J GavardJS GutkindVEGF controls endothelial cell permeability by promoting the beta-arrestin-dependent endocytosis of VE-cadherinNat Cell Biol812231234200610.1038/ncb148617060906

23. 

T AsaharaT TakahashiH MasudaVEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cellsEMBO J1839643972199910.1093/emboj/18.14.396410406801

24. 

C KalkaH MasudaT TakahashiVascular endothelial growth factor (165) gene transfer augments circulating endothelial progenitor cells in human subjectsCirc Res8611981202200010.1161/01.RES.86.12.1198

25. 

M GillS DiasK HattoriML RiveraD HicklinL WitteL GirardiR YurtH HimelS RafiiVascular trauma induces rapid but transient mobilization of VEGFR2(+)AC133(+) endothelial precursor cellsCirc Res88167174200111157668

26. 

F BautzS RafiiL KanzR MohleExpression and secretion of vascular endothelial growth factor-A by cytokine stimulated hematopoietic progenitor cells: possible role in the hematopoietic microenvironmentExp Hematol28700706200010.1016/S0301-472X(00)00168-5

27. 

TM PowellJD PaulJM HillGranulocyte colony-stimulating factor mobilizes functional endothelial progenitor cells in patients with coronary artery diseaseArterioscler Thromb Vasc Biol25296301200510.1161/01.ATV.0000151690.43777.e4

28. 

B HeissigK HattoriS DiasM FriedrichB FerrisNR HackettRecruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligandCell109625637200210.1016/S0092-8674(02)00754-712062105

29. 

DJ CeradiniAR KulkarniMJ CallaghanOM TepperN BastidasME KleinmanProgenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1Nat Med10858864200410.1038/nm107515235597

30. 

E De FalcoD PorcelliAR TorellaS StrainoMG IachininotoA OrlandiS TruffaP BiglioliM NapolitanoMC CapogrossiM PesceSDF-1 involvement in endothelial phenotype and ischemia-induced recruitment of bone marrow progenitor cellsBlood10434723482200415284120

31. 

K HattoriB HeissigK TashiroPlasma elevation of stromal cell-derived factor-1 induces mobilization of mature and immature hematopoietic progenitor and stem cellsBlood9733543360200110.1182/blood.V97.11.335411369624

32. 

DH WalterJ HaendelerJ ReinholdU RochwalskyF SeegerJ HonoldJ HoffmannC UrbichR LehmannF Arenzana-SeisdesdosImpaired CXCR4 signaling contributes to the reduced neovascularization capacity of endothelial progenitor cells from patients with coronary artery diseaseCirc Res9711421151200510.1161/01.RES.0000193596.94936.2c

33. 

T AsaharaJM IsnerEndothelial progenitor cells for vascular regenerationJ Hematother Stem Cell Res11171178200210.1089/15258160275365838511983091

34. 

O KolletS ShivtielYQ ChenJ SuriawinataSN ThungMD DabevaHGF, SDF-1, and MMP-9 are involved in stress-induced human CD34+ stem cell recruitment to the liverJ Clin Invest112160169200310.1172/JCI1790212865405

35. 

AT AskariS UnzekZB PopovicCK GoldmanF ForudiM KiedrowskiEffect of stromal-cell-derived factor 1 on stem cell homing and tissue regeneration in ischaemic cardiomyopathyLancet362697703200310.1016/S0140-6736(03)14232-812957092

36. 

H LangerAE MayK DaubU HeinzmannP LangM SchummD VestweberS MassbergT SchonbergerI PfistererAdherent platelets recruit and induce differentiation of murine embryonic endothelial progenitor cells to mature endothelial cells in vitroCirc Res98e2e10200610.1161/01.RES.0000201285.87524.9e

37. 

C UrbichA AicherC HeeschenE DernbachWK HofmannAM ZeiherS DimmelerSoluble factors released by endothelial progenitor cells promote migration of endothelial cells and cardiac resident progenitor cellsJ Mol Cell Cardiol39733742200510.1016/j.yjmcc.2005.07.00316199052

38. 

J ChenJA LopezInteractions of platelets with subendothelium and endotheliumMicrocirculation12235246200510.1080/1073968059092548415814433

39. 

ZM RuggeriMechanisms initiating platelet thrombus formationThromb Haemost7861161619979198225

40. 

S MassbergM GawazS GrunerV SchulteI KonradD ZohlnhoferA crucial role of glycoprotein VI for platelet recruitment to the injured arterial wall in vivoJ Exp Med1974149200310.1084/jem.2002094512515812

41. 

S MassbergK SchurzingerM LorenzI KonradC SchulzN PlesnilaE KennerknechtM RudeliusS SauerS BraunPlatelet adhesion via glycoprotein IIb integrin is critical for atheroprogression and focal cerebral ischemia: an in vivo study in mice lacking glycoprotein IIbCirculation11211801188200510.1161/CIRCULATIONAHA.105.53922116103235

42. 

S MassbergI KonradK SchurzingerM LorenzS SchneiderD ZohlnhoeferPlatelets secrete stromal cell-derived factor1alpha and recruit bone marrow-derived progenitor cells to arterial thrombi in vivoJ Exp Med20312211233200610.1084/jem.2005177216618794

43. 

K HiasaM IshibashiK OhtaniS InoueQ ZhaoS KitamotoGene transfer of stromal cell-derived factor-1alpha enhances ischemic vasculogenesis and angiogenesis via vascular endothelial growth factor/endothelial nitric oxide synthase related pathway: next-generation chemokine therapy for therapeutic neovascularizationCirculation109245424612004

44. 

A AicherC HeeschenC Mildner-RihmC UrbichC IhlingK Technau-IhlingAM ZeiherS DimmelerEssential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cellsNat Med913701376200310.1038/nm94814556003

45. 

L LaterveerIJ LindleyMS HamiltonR WillemzeWE FibbeInterleukin-8 induces rapid mobilization of hematopoietic stem cells with radioprotective capacity and long term myelolymphoid repopulating abilityBlood852269227519957718900

46. 

L LaterveerJM ZijlemansIJ LindleyMS HamiltonR WillemzeWE FibbeImproved survival of lethally irradiated recipient mice transplanted with circulating progenitor cells mobilized by IL-8 after pretreatment with stem cell factorExp Hematol241387139319968913284

47. 

RM PalmerAG FerrigeS MoncadaNitric oxide release accounts for the biological activity of endothelium-derived relaxing factorNature327524526198710.1038/327524a03495737

48. 

JA WareDD HeistadSeminars in medicine of the Beth Israel Hospital, Boston. Platelet-endothelium interactionsN Engl J Med328628635199310.1056/NEJM1993030432809078429855

49. 

LJ GoldsteinKA GallagherSM BauerRJ BauerV BaireddyZJ LiuDG BuerkSR ThomOC VelazquezEndothelial progenitor cell release into circulation is triggered by hyperoxia-induced increases in bone marrow nitric oxideStem Cells2423092318200610.1634/stemcells.2006-001016794267

50. 

SR ThomVM BhopaleOC VelazquezLJ GoldsteinLH ThomDG BuerkStem cell mobilization by hyperbaric oxygenAm J Physiol Heart Circ Physiol290H1378H1386200610.1152/ajpheart.00888.200516299259

51. 

OW GriffithDJ StuehrNitric oxide synthases: properties and catalytic mechanismAnnu Rev Physiol57707736199510.1146/annurev.ph.57.030195.0034237539994

52. 

MA MarlettaAR HurshmanKM RuscheCatalysis by nitric oxide synthaseCurr Opin Chem Biol2656663199810.1016/S1367-5931(98)80098-7

53. 

Z LaczaM PuskarJP FigueroaJ ZhangN RajapakseDW BusijaMitochondrial nitric oxide synthase is constitutively active and is functionally upregulated in hypoxiaFree Radic Biol Med3116091615200110.1016/S0891-5849(01)00754-711744335

54. 

KA GallagherZL LiuM XiaoH ChenDiabetic impairments in NO-mediated endothelial progenitor cell mobilization and homing are reversed by hyperoxia and SDF-1αJ Clin Invest11712491259200717476357

55. 

SK GuptaPG LyskoK PillarisettiE OhlsteinJM StadelChemokine receptors in human endothelial cells. Functional expression of CXCR4 and its transcriptional regulation by inflammatory cytokinesJ Biol Chem27342824287199810.1074/jbc.273.7.42829461627

56. 

S BhaktaP HongO KocThe surface adhesion molecule CXCR4 stimulates mesenchymal stem cell migration to stromal cell-derived factor-1 in vitro but does not decrease apoptosis under serum deprivationCardiovasc Revasc Med71924200610.1016/j.carrev.2005.10.008

57. 

M KuciaR RecaK MiekusJ WanzeckW WojakowskiA Janowska-WieczorekJ RatajczakMZ RatajczakTrafficking of normal stem cells and metastasis of cancer stem cells involve similar mechanisms: pivotal role of the SDF-1-CXCR4 axisStem Cells23879894200510.1634/stemcells.2004-034215888687

58. 

T LapidotMechanism of human stem cell migration and repopulation of NOD/SCID and B2m Null NOD/SCID mouse: the role of SDF-1/CXCR4 interactionsAnn NY Acad Sci9388395200110.1111/j.1749-6632.2001.tb03577.x11458529

59. 

H ZhengG FuT DaiH HuangMigration of endothelial progenitor cells mediated by stromal cell-derived factor-1alpha/CXCR4 via PI3K/Akt/eNOS signal transduction pathwayJ Cardiovasc Pharmacol50274280200710.1097/FJC.0b013e318093ec8f17878755

60. 

S AkiraT TagaT KishimotoInterleukin-6 in biology and medicineAdv Immunol54178199310.1016/S0065-2776(08)60532-5

61. 

M HibiK NakajimaT HiranoIL-6 cytokine family and signal transduction: a model of the cytokine systemJ Mol Med74112199610.1007/BF002020688834766

62. 

D KamimuraK IshiharaT HiranoTransduction and its physiological roles: the signal orchestration modelRev Physiol Biochem Pharmacol149138200310.1007/s10254-003-0012-212687404

63. 

MB NilssonRR LangleyIJ FidlerInterleukin-6, secreted by human ovarian carcinoma cells, is a potent proangiogenic cytokineCancer Res651079410800200510.1158/0008-5472.CAN-05-062316322225

64. 

Z WangWH NewmanSmooth muscle cell migration stimulated by interleukin-6 is associated with cytoskeletal reorganizationJ Surg Res111261266200310.1016/S0022-4804(03)00087-812850472

65. 

JS YaoW ZhaiWL YoungGY YangInterleukin-6 triggers human cerebral endothelial cells proliferation and migration: the role for KDR and MMP-9Biochem Biophys Res Commun34213961404200610.1016/j.bbrc.2006.02.10016516857

66. 

Y FanY YeF ShenInterleukin-6 stimulates circulating blood-derived endothelial progenitor cell angiogenesis in vitroJ Cereb Blood Flow Metab289098200810.1038/sj.jcbfm.960050917519976

67. 

M BartoliD PlattT LemtalsiX GuSE BrooksMB MarreroRB CaldwellVEGF differentially activates STAT3 in microvascular endothelial cellsFASEB J1715621564200312824281

68. 

DD DeoTW AxelradEG RobertV MarcheselliNG BazanJD HuntPhosphorylation of STAT-3 in response to basic fibroblast growth factor occurs through a mechanism involving platelet-activating factor, JAK-2, and Src in human umbilical vein endothelial cells. Evidence for a dual kinase mechanismJ Biol Chem2772123721245200210.1074/jbc.M110955200

69. 

IU SchraufstatterJ ChungM BurgerIL-8 activates endothelial cell CXCR1 and CXCR2 through Rho and Rac signaling pathwaysAm J Physiol Lung Cell Mol Physiol280L1094L1103200111350788

70. 

C HeissML WongWI BlockD LaoMW RealY YeghiazariansRL LeeML SpringerPleiotrophin induces nitric oxide dependent migration of endothelial progenitor cellsJ Cell Physiol215366373200810.1002/jcp.2131317960557

71. 

TD PollardGG BorisyCellular motility driven by assembly and disassembly of actin filamentsCell112453465200310.1016/S0092-8674(03)00120-X12600310

72. 

C RevenuR AthmanS RobineD LouvardThe coworkers of actin filaments: from cell structures to signalsNat Rev Mol Cell Biol5635646200410.1038/nrm143715366707

73. 

J WeiY LiuM ChangCL SunDW LiZQ LiuLS HuProteomic analysis of oxidative modification in endothelial colony-forming cells treated by hydrogen peroxideInt J Mol Med2910991105201222446847

74. 

N WernerJ PrillerU LaufsM EndresM BohmU DirnaglG NickenigBone marrow-derived progenitor cells modulate vascular reendothelialization and neointimal formation: effect of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitionArterioscler Thromb Vasc Biol2215671572200210.1161/01.ATV.0000036417.43987.D8

75. 

M SataA SaiuraA KunisatoA TojoS OkadaT TokuhisaH HiraiM MakuuchiY HirataR NagaiHematopoietic stem cells differentiate into vascular cells that participate in the pathogenesis of atherosclerosisNat Med8403409200210.1038/nm0402-40311927948

76. 

D SimperPG StalboergerCL PanettaS WangNM CapliceSmooth muscle progenitor cells in human bloodCirculation10611991204200210.1161/01.CIR.0000031525.61826.A812208793

77. 

K ShimizuS SugiyamaM AikawaY FukumotoE RabkinP LibbyRN MitchellHost bone-marrow cells are a source of donor intimal smooth-muscle-like cells in murine aortic transplant arteriopathyNat Med7738741200111385513

78. 

JD AbbottY HuangD LiuR HickeyDS KrauseFJ GiordanoStromal cell-derived factor-1alpha plays a critical role in stem cell recruitment to the heart after myocardial infarction but is not sufficient to induce homing in the absence of injuryCirculation11033003305200410.1161/01.CIR.0000147780.30124.CF15533866

79. 

A SchoberS KnarrenM LietzEA LinC WeberCrucial role of stromal cell-derived factor-1alpha in neointima formation after vascular injury in apolipoprotein E deficient miceCirculation10824912497200310.1161/01.CIR.0000099508.76665.9A14581398

80. 

A ZerneckeA SchoberI BotP von HundelshausenEA LiehnB MoppsM MericskayP GierschikEA BiessenC WeberSDF-1alpha/CXCR4 axis is instrumental in neointimal hyperplasia and recruitment of smooth muscle progenitor cellsCirc Res96784791200510.1161/01.RES.0000162100.52009.3815761195

81. 

E ChavakisA AicherC HeeschenRole of beta2-integrins for homing and neovascularization capacity of endothelial progenitor cellsJ Exp Med2016372200510.1084/jem.2004140215623573

82. 

E ChavakisA HainM VinciG CarmonaME BianchiP VajkoczyHigh mobility group box1 activates integrin dependent homing of endothelial progenitor cellsCirc Res100204212200710.1161/01.RES.0000257774.55970.f417218606

83. 

B DelormeA BasireC GentileF SabatierF MonsonisC DesouchesPresence of endothelial progenitor cells, distinct from mature endothelial cells, within human CD146+ blood cellsThromb Haemost9412701279200516411405

84. 

T BombeliBR SchwartzJM HarlanAdhesion of activated platelets to endothelial cells: evidence for a GPIIbIIIa-dependent bridging mechanism and novel roles for endothelial intercellular adhesion molecule 1 (ICAM-1), alphavbeta3 integrin, and GPIbalphaJ Exp Med187329339199810.1084/jem.187.3.329

85. 

H JinA AiyerJ SuP BorgstromD StupackM FriedlanderJ VarnerA homing mechanism for bone marrow-derived progenitor cell recruitment to the neovasculatureJ Clin Invest116652662200610.1172/JCI2475116498499

86. 

G QinM IiM SilverA WeckerE BordH MaFunctional disruption of alpha4 integrin mobilizes bone marrow derived endothelial progenitors and augments ischemic neovascularizationJ Exp Med203153163200610.1084/jem.20050459

87. 

A KawamotoHC GwonH IwaguroJI YamaguchiS UchidaH MasudaM SilverH MaM KearneyJM IsnerT AsaharaTherapeutic potential of ex vivo expanded endothelial progenitor cells for myocardial ischemiaCirculation103634637200110.1161/01.CIR.103.5.63411156872

88. 

C UrbichC HeeschenA AicherE DernbachAM ZeiherS DimmelerRelevance of monocytic features for neovascularization capacity of circulating endothelial progenitor cellsCirculation10825112516200310.1161/01.CIR.0000096483.29777.5014581410

89. 

A KawamotoT TkebuchavaJ YamaguchiH NishimuraYS YoonC MillikenS UchidaO MasuoH IwaguroH MaIntramyocardial transplantation of autologous endothelial progenitor cells for therapeutic neovascularization of myocardial ischemiaCirculation107461468200310.1161/01.CIR.0000046450.89986.50

90. 

T MuroharaH IkedaJ DuanS ShintaniK SasakiH EguchiI OnitsukaK MatsuiT ImaizumiTransplanted cord blood derived endothelial precursor cells augment postnatal neovascularizationJ Clin Invest10515271536200010.1172/JCI829610841511

91. 

E Tateishi-YuyamaH MatsubaraT MuroharaU IkedaS ShintaniH MasakiK AmanoY KishimotoK YoshimotoH AkashiTherapeutic angiogenesis for patients with limb ischaemia by autologous transplantation of bone-marrow cells: a pilot study and a randomised controlled trialLancet360427435200210.1016/S0140-6736(02)09670-8

92. 

B AssmusV SchächingerC TeupeM BrittenR LehmannN DöbertF GrünwaldA AicherC UrbichH MartinD HoelzerS DimmelerAM ZeiherTransplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (TOPCARE-AMI)Circulation10630093017200210.1161/01.CIR.0000043246.74879.CD

93. 

JC KovacicJ MooreA HerbertD MaM BoehmRM GrahamEndothelial progenitor cells, angioblasts, and angiogenesis: old terms reconsidered from a current perspectiveTrends Cardiovasc Med184551200810.1016/j.tcm.2007.12.00218308194

94. 

KK HirschiDA IngramMC YoderAssessing identity, phenotype, and fate of endothelial progenitor cellsArterioscler Thromb Vasc Biol2815841595200810.1161/ATVBAHA.107.15596018669889

95. 

CE MurryMH SoonpaaH ReineckeHaematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarctsNature428664668200410.1038/nature0244615034593

96. 

LB BalsamAJ WagersJL ChristensenT KofidisIL WeissmanRC RobbinsHaematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardiumNature428668673200410.1038/nature0246015034594

97. 

E KamoshitaY IkedaM FujitaH AmanoA OikawaRecruitment of a prostaglandin E receptor subtype, EP3-expressing bone marrow cells is crucial in wound-induced angiogenesisAm J Pathol16914581472200610.2353/ajpath.2006.05135817003499

Related Articles

Journal Cover

November 2012
Volume 30 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li D, Liu Z, Wei J, Liu Y and Hu L: Contribution of endothelial progenitor cells to neovascularization (Review). Int J Mol Med 30: 1000-1006, 2012
APA
Li, D., Liu, Z., Wei, J., Liu, Y., & Hu, L. (2012). Contribution of endothelial progenitor cells to neovascularization (Review). International Journal of Molecular Medicine, 30, 1000-1006. https://doi.org/10.3892/ijmm.2012.1108
MLA
Li, D., Liu, Z., Wei, J., Liu, Y., Hu, L."Contribution of endothelial progenitor cells to neovascularization (Review)". International Journal of Molecular Medicine 30.5 (2012): 1000-1006.
Chicago
Li, D., Liu, Z., Wei, J., Liu, Y., Hu, L."Contribution of endothelial progenitor cells to neovascularization (Review)". International Journal of Molecular Medicine 30, no. 5 (2012): 1000-1006. https://doi.org/10.3892/ijmm.2012.1108