SlideShare a Scribd company logo
Endothelial Progenitor Cells : Characterization and Role in Vascular Biology
                     Carmen Urbich and Stefanie Dimmeler


                           Circulation Research 2004, 95:343-353
                         doi: 10.1161/01.RES.0000137877.89448.78
Circulation Research is published by the American Heart Association. 7272 Greenville Avenue, Dallas,
                                             TX 72514
  Copyright © 2004 American Heart Association. All rights reserved. Print ISSN: 0009-7330. Online
                                          ISSN: 1524-4571




  The online version of this article, along with updated information and services, is
                         located on the World Wide Web at:
                            http://circres.ahajournals.org/content/95/4/343




  Subscriptions: Information about subscribing to Circulation Research is online at
  http://circres.ahajournals.org//subscriptions/

  Permissions: Permissions & Rights Desk, Lippincott Williams & Wilkins, a division of Wolters
  Kluwer Health, 351 West Camden Street, Baltimore, MD 21202-2436. Phone: 410-528-4050. Fax:
  410-528-8550. E-mail:
  journalpermissions@lww.com

  Reprints: Information about reprints can be found online at
  http://www.lww.com/reprints




        Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
Reviews

     This Review is part of a thematic series on Angiogenesis, which includes the following articles:

     Endothelial Progenitor Cells: Characterization and Role in Vascular Biology

     Bone Marrow–Derived Cells for Enhancing Collateral Development: Mechanisms, Animal Data, and Initial Clinical
     Experiences
     Arteriogenesis
     Innate Immunity and Angiogenesis
     Syndecans
     Growth Factors and Blood Vessels: Differentiation and Maturation                                            Ralph Kelly, Guest Editor




                                       Endothelial Progenitor Cells
                           Characterization and Role in Vascular Biology
                                                Carmen Urbich, Stefanie Dimmeler

Abstract—Infusion of different hematopoietic stem cell populations and ex vivo expanded endothelial progenitor cells
  augments neovascularization of tissue after ischemia and contributes to reendothelialization after endothelial injury,
  thereby, providing a novel therapeutic option. However, controversy exists with respect to the identification and the
  origin of endothelial progenitor cells. Overall, there is consensus that endothelial progenitor cells can derive from the
  bone marrow and that CD133/VEGFR2 cells represent a population with endothelial progenitor capacity. However,
  increasing evidence suggests that there are additional bone marrow– derived cell populations (eg, myeloid cells, “side
  population” cells, and mesenchymal cells) and non-bone marrow– derived cells, which also can give rise to endothelial
  cells. The characterization of the different progenitor cell populations and their functional properties are discussed.
  Mobilization and endothelial progenitor cell–mediated neovascularization is critically regulated. Stimulatory (eg, statins
  and exercise) or inhibitory factors (risk factors for coronary artery disease) modulate progenitor cell levels and, thereby,
  affect the vascular repair capacity. Moreover, recruitment and incorporation of endothelial progenitor cells requires a
  coordinated sequence of multistep adhesive and signaling events including adhesion and migration (eg, by integrins),
  chemoattraction (eg, by SDF-1/CXCR4), and finally the differentiation to endothelial cells. This review summarizes the
  mechanisms regulating endothelial progenitor cell–mediated neovascularization and reendothelialization. (Circ Res.
  2004;95:343-353.)
          Key Words: progenitor cells Ⅲ neovascularization Ⅲ vasculogenesis Ⅲ angiogenesis Ⅲ endothelial cells



D     ifferentiation of mesodermal cells to angioblasts and
      subsequent endothelial differentiation was believed to
exclusively occur in embryonic development. This dogma
                                                                             mia. Rafii’s group in 19982 also reported the existence of
                                                                             “circulating bone marrow– derived endothelial progenitor
                                                                             cells” (CEPCs) in the adult. Again, a subset of CD34ϩ
was overturned in 1997, when Asahara and colleagues1                         hematopoietic stem cells was shown to differentiate to the
published that purified CD34ϩ hematopoietic progenitor cells                 endothelial lineage and express endothelial marker proteins
from adults can differentiate ex vivo to an endothelial                      such as vWF and incorporated Dil-Ac-LDL. Most convinc-
phenotype. These cells were named “endothelial progenitor                    ingly, bone marrow–transplanted genetically tagged cells
cells” (EPCs), showed expression of various endothelial                      were covering implanted Dacron grafts.2 These pioneering
markers, and incorporated into neovessels at sites of ische-                 studies suggested the presence of circulating hemangioblasts


  Original received March 8, 2004; revision received May 27, 2004; accepted May 28, 2004.
  From Molecular Cardiology, Department of Internal Medicine IV, University of Frankfurt, Frankfurt, Germany.
  Correspondence to Stefanie Dimmeler, PhD, Molecular Cardiology, Dept of Internal Medicine IV, University of Frankfurt, Theodor-Stern-Kai 7, 60590
Frankfurt, Germany. E-mail Dimmeler@em.uni-frankfurt.de
  © 2004 American Heart Association, Inc.
  Circulation Research is available at http://www.circresaha.org                                      DOI: 10.1161/01.RES.0000137877.89448.78

                                                                  343
                     Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
344      Circulation Research           August 20, 2004


in the adult. According to the initial discovery, EPCs or            endothelial lineage. Interestingly, lineage tracking showed
CEPCs were defined as cells positive for both hematopoi-             that myeloid cells are the hematopoietic stem cell– derived
etic stem cell markers such as CD34 and an endothelial               intermediates, which contribute to muscle regeneration,10
marker protein as VEGFR2. Because CD34 is not exclu-                 suggesting that myeloid intermediates may be part of the
sively expressed on hematopoietic stem cells but, albeit at          repair capacity after injury. Moreover, a subset of human
a lower level, also on mature endothelial cells, further             peripheral blood monocytes acts as pluripotent stem cells.11
studies used the more immature hematopoietic stem cell                  Of note, a specific problem arises when cells are ex vivo
marker CD1333 and demonstrated that purified CD133ϩ                  expanded and cultured, because the culture conditions (cul-
cells can differentiate to endothelial cells in vitro.4 CD133,       ture supplements such as FCS and cytokines, plastic) rapidly
also known as prominin or AC133, is a highly conserved               changes the phenotype of the cells. For example, supplemen-
antigen with unknown biological activity, which is ex-               tation of the medium with statins increased the number of
pressed on hematopoietic stem cells but is absent on                 endothelial cell colonies isolated from mononuclear cells.12
mature endothelial cells and monocytic cells (see review).5          Moreover, continuous cultivation was shown to increase
Thus, CD133ϩVEGFR2ϩ cells more likely reflect imma-                  endothelial marker protein expression.13 This may explain
ture progenitor cells, whereas CD34ϩVEGFR2ϩ may also                 why different groups may obtain cells with different surface
represent shedded cells of the vessel wall. At present, it is        factor profile and functional activity although similar proto-
unclear whether CD133 only represents a surface marker               cols were used for cultivation.9,14 –16 Moreover, the interaction
or has a functional activity involved in regulation of               of cells within a heterogeneous mixture of cells such as the
neovascularization.                                                  mononuclear cells from the blood may impact the yield and
   Overall, controversy exists with respect to the identifica-       the functional activity of the cultivated cells.17
tion and the origin of endothelial progenitor cells, which are          Generally, several studies suggested that other cell popu-
isolated from peripheral blood mononuclear cells by cultiva-         lations beside hematopoietic stem cells also can give rise to
tion in medium favoring endothelial differentiation. In pe-          endothelial cells (Figure 1). Thus, non-bone marrow– derived
ripheral blood mononuclear cells, several possible sources for       cells have been shown to replace the endothelial cells in
endothelial cells may exist: (1) the rare number of hemato-          grafts.18 In addition, adult bone marrow– derived stem/pro-
poietic stem cells, (2) myeloid cells, which may differentiate       genitor cells such as the side population cells and multipotent
to endothelial cells under the cultivation selection pressure,       adult progenitor cells, which are distinct from hematopoietic
(3) other circulating progenitor cells (eg, “side population”        stem cells, have also been shown to differentiate to the
cells), and (4) circulating mature endothelial cells, which are      endothelial lineage.19,20 Recently, tissue-resident stem cells
shed off the vessel wall6 and adhere to the culture dishes. First    have been isolated from the heart, which are capable to
evidence that there is more than one endothelial progeny             differentiate to the endothelial lineage.21 These data support
within the circulating blood was provided by Hebbel and              the notion that it will be difficult to define the “true”
colleagues, who showed that morphological and functional             endothelial progenitor cells. Overall, the field is reminiscent
distinct endothelial cell populations can be grown out of            to immunology, where T-cells initially were defined as one
peripheral blood mononuclear cells.7 They stratified the             cell population. However, the functional characterization (eg,
different circulating endothelial cells according to their           cytokine release and response to stimuli) helped to identify
growth characteristics and morphological appearance as               novel T-cell subpopulations with distinct functions and ca-
“spindle-like cells,” which have a low proliferative capacity,       pacities. Hopefully, better profiling of distinct cell popula-
and outgrowing cells. Because the outgrowing cells showed a          tions and fate mapping studies will help to identify markers,
high proliferative potential and originated predominantly            which distinguish the circulating endothelial precursor within
from the bone marrow donors, they were considered as circu-          the blood and bone marrow/non-bone marrow– derived endo-
lating angioblasts.7 The authors speculated that the spindle-like    thelial cells.
cells may likely represent mature endothelial cells, which are
shed off the vessel wall. However, this hypothesis is difficult to          Role of EPCs in Neovascularization
test and has not yet been proven thus far.                           Improvement of neovascularization is a therapeutic option to
   Experimentally, preplating may be a way to reduce the             rescue tissue from critical ischemia.22 The finding that bone
heterogeneity of the cultivated EPCs, because this excludes          marrow– derived cells can home to sites of ischemia and
rapidly adhering cells such as differentiated monocytic or           express endothelial marker proteins has challenged the use of
possible mature endothelial cells.2 However, these protocols         isolated hematopoietic stem cells or EPCs for therapeutic
do not eliminate myeloid and nonhematopoietic progenitor             vasculogenesis. Infusion of various distinct cell types either
cells, which may contribute to the ex vivo cultivated cells.         isolated from the bone marrow or by ex vivo cultivation was
There is increasing evidence that myeloid cells can give rise        shown to augment capillary density and neovascularization of
to endothelial cells as well. Specifically, CD14ϩ/CD34Ϫ              ischemic tissue (Table 1 and Figure 2). In animal models of
myeloid cells can coexpress endothelial markers and form             myocardial infarction, the injection of ex vivo expanded
tube-like structures ex vivo.8 Additionally, ex vivo expansion       EPCs or stem and progenitor cells significantly improved
of purified CD14ϩ mononuclear cells yielded cells with an            blood flow and cardiac function and reduced left ventricular
endothelial characteristic, which incorporated in newly              scarring.23,24 Similarly, infusion of ex vivo expanded EPCs
formed blood vessels in vivo.9 These data would suggest that         deriving from peripheral blood mononuclear cells in nude
myeloid cells can differentiate (or transdifferentiate) to the       mice or rats improved the neovascularization in hind limb
                    Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
Urbich and Dimmeler                Endothelial Progenitor Cells and Vascular Biology                345




                                                                                               Figure 1. Origin and differentiation of
                                                                                               endothelial progenitor cells. Scheme
                                                                                               depicts the potential origin and differenti-
                                                                                               ation of endothelial progenitor cells from
                                                                                               hematopoietic stem cells and nonhema-
                                                                                               topoietic cells.




ischemia models.9,15,23,25 Correspondingly, initial pilot trials         uncultivated CD34Ϫ cells. Remarkably, terminally differenti-
indicate that bone marrow– derived or circulating blood–                 ated mature endothelial cells (HMVECs, GEAECs, and
derived progenitor cells are useful for therapeutically improv-          SVECs) did not improve neovascularization15,24,33 suggesting
ing blood supply of ischemic tissue.26,27 Autologous implan-             that a not-yet-defined functional characteristic (eg, chemo-
tation of bone marrow mononuclear cells in patients with                 kine or integrin receptors mediating homing) is essential for
ischemic limbs significantly augmented ankle-brachial index              EPC-mediated augmentation of blood flow after ischemia.
and reduced rest pain.26 In addition, transplantation of ex vivo            The functional capacity of EPCs to augment blood flow
expanded endothelial progenitor cells significantly improved             further does not appear to be solely attributable to a mono-
coronary flow reserve and left ventricular function in patients          cytic phenotype. Ex vivo cultivated EPCs from CD14ϩ
with acute myocardial infarction.27                                      mononuclear cells or CD14Ϫ mononuclear cell starting pop-
   Besides models of peripheral ischemia (hind limb ische-               ulation improved neovascularization to a similar extent,
mia), the angiogenic potential of EPCs was also investigated             whereas the same number of freshly isolated mononuclear
in animal models of tumor angiogenesis. Thereby, the inhi-               cells taken from the starting culture did not.9 Interestingly,
bition of VEGF-responsive bone marrow– derived endothelial               these experimental data are supported by first clinical trials
and hematopoietic precursor cells blocks tumor angiogenesis              showing that freshly isolated mononuclear cells are not well
and growth.28 The use of various different models, cell                  suited to improve neovascularization in patients with periph-
numbers, and species limits the comparability of the effi-               eral vascular diseases.26 However, monocytic cells may play
ciency of distinct cell populations. However, the overall                a crucial role in collateral growth (arteriogenesis). Thus, the
functional improvement appear similar, when isolated human               attraction of monocytic cells by monocyte chemoattractant
CD34ϩ, CD133ϩ, EPC, MAPC, or murine Sca-1ϩ cells were                    protein-1 (MCP-1) enhanced arteriogenesis.34 Moreover, de-
used.4,9,15,20,23,25,29 –32 Likewise, early spindle-like cells and       pletion of the monocytes reduced PlGF-induced arteriogen-
late outgrowing EPCs showed comparable in vivo vasculo-                  esis.35 A therapeutic benefit of monocyte infusion on arterio-
genic capacity.33 These results suggest that the functional              genesis was demonstrated under conditions of monocyte
activity of the cells to augment neovascularization is rather            deficiency induced by chemical depletion.36 These data sug-
independent of the type of (endothelial) progenitor cell used.           gest that monocytic cells are necessary for arteriogenesis and
However, the CD34Ϫ fractions of freshly isolated bone                    possibly neovascularization. For therapeutic application, the
marrow– or blood-derived mononuclear cells showed a re-                  local enhancement of monocyte recruitment might be better
duced incorporation and functional activity.24,29 These data             suited than systemic infusion of monocytic cells, which only
indicate that the number of cells capable to augment neovas-             leads to a relatively minor increase in the number of circu-
cularization is low in this crude fraction of freshly isolated           lating monocytes.
                    Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
346             Circulation Research             August 20, 2004


TABLE 1.           Neovascularization Induced by Injection of Progenitor Cells: Experimental and Clinical Studies
Cells                                              Surface Markers                                     Improvement Models                               Incorporation Rate
Experimental studies
  Freshly isolated cells
        CD34ϩ cells                             CD34ϩ/flk-1ϩ, CD45ϩ 1                                      Incorporation1                      13.4 Ϯ5.7% (mouse) or 9.7 Ϯ4.5%
                                                                                                                                                (rabbit) Dil-Ac-LDL-EPC in CD31ϩ
                                                                                                                                                             capillaries1
                                                Tie-2ϩ, Dil-Ac-LDLϩ   29
                                                                                                       Hind limb ischemia29                          Frequently detected (not
                                                                                                                                                          quantified)29
                                       CD117bright/GATAϪ2/VEGFR2/Tie-2/AC133   24
                                                                                                       Myocardial infarction24                 20–25% of total myocardial capillary
                                                                                                                                                         vasculature24
        Sca-1ϩ BM-MNCs                                 Sca-1ϩ 30                                       Hind limb ischemia30                          Detected (not quantified)
        PBMCs                        T and B lymphocytes and monocytes-depleted                        Hind limb ischemia30
                                                       MNCs30
  Ex vivo expanded cells
        Ex vivo expanded EPC          Dil-Ac-LDLϩ/lectinϩ VEGFR2ϩ, VE-cadherinϩ,                      Hind limb ischemia15,31                    2.1 Ϯ0.4 EPCs into vessels in a
                                              CD31ϩ, CD14ϩ, CD34ϩ 15,23                               Myocardial infarction23                             ϫ10 field15
                                                                                                                                                 241 Ϯ25 cells/mm2 (day 3) 355
                                                                                                                                                     Ϯ30 cells/mm2 (day 7)31
                                       Dil-Ac-LDLϩ, NOϩ, VEGFR2ϩ, VE-cadherinϩ,                        Hind limb ischemia25                          Frequently detected (not
                                                CD31ϩ, vWFϩ, CD45Ϫ 25                                                                                     quantified)25

                                     CD31ϩ, vWFϩ, Dil-Ac-LDLϩ, VEGFR2ϩ, Tie-2ϩ      53
                                                                                          Vascular graft survival, Neovessel remodeling53        80% of graft lumen at day 1553

                                                 Dil-Ac-LDLϩ/lectinϩ                                    Hind limb ischemia9                     19.8 Ϯ8% CD146ϩ/HLA-DRϩ cell
                                           VEGFR2ϩ, CD105ϩ, vWFϩ, CD45ϩ    9
                                                                                                                                                      containing vessels9

                                    Early EPC: Dil-Ac-LDLϩ/lectinϩ VEGFR2ϩ, CD31ϩ,                     Matrigel capillaries16                                  ND
                                         Tie-2ϩ, VE-cadherinϪ, eNOSϪ, CD14ϩ 16           Outgrowing ECs: exhibited a greater capacity for
                                       Outgrowing ECs: VEGFR2ϩ, CD31ϩ, Tie-2ϩ,            capillary morphogenesis in in vitro and in vivo
                                             VE-cadherinϩ, eNOSϩ, CD14Ϫ 16                               matrigel models

                                          Early EPC: weak VEGFR1, eNOS, vWF,             In vitro: late EPC showed better incorporation and         Detected (not quantified)33
                                          VE-cadherin, VEGFR2, spindle shape33              tube formation. Early EPC: higher release of
                                     Late EPC: strong VE-cadherin, VEGFR1, VEGFR2,        growth factors. In vivo: comparable vasculogenic
                                          eNOS, vWF, cobblestone morphology33              potential of early and late EPC (limb perfusion,
                                                                                                           capillary density)

        MAPC-derived ECs                Co-purified MAPC: CD34Ϫ, VE-cadherinϪ,                     tumor growth/angiogenesis20                         MAPC-derived ECs20
                                                    AC133ϩ, Flk-1ϩ 20                                                                           35% tumor angiogenesis, 30–45%
                                       Angioblast: CD34ϩ, VE-cadherinϩ, AC133Ϫ,
                                                              ϩ 20
                                                                                                                                                  wound healing angiogenesis,
                                                        Flk-1                                                                                     undifferentiated MAPCs: 12%
Clinical studies
  BMC and monocytes (TACT-trial)              CD34ϩ/Dil-Ac-LDLϩ/lectinϩ                  Intramuscular injection in patients with peripheral                   ND
                                                                                              ischemic disease; improved blood flow26
  CPC and BMC (TOPCARE-AMI)           CPC: Dil-Ac-LDLϩ/lectinϩ, VEGFR2ϩ, CD31ϩ,             Intracoronary infusion in patients with AMI;                       ND
                                         vWFϩ, CD105ϩ; BMC: CD34ϩ/CD45ϩ,                         increase in coronary flow reserve27
                                           CD34ϩ/CD133ϩ, CD34ϩ/VEGFR2ϩ



Mechanisms by Which EPC                                                                     derived cells adjacent to vessels, which do not express
Improve Neovascularization                                                                  endothelial marker proteins.41,45 A reasonable explanation
Although the role of EPCs in neovascularization has been                                    might be that the model of ischemia (eg, intensity of injury or
convincingly shown by several groups, the question remains:                                 ischemia)46 significantly influences the incorporation rate. A
how do EPCs improve neovascularization?                                                     minor ischemia might not as profoundly induce a mobiliza-
   Bone marrow transplantation of genetically modified cells                                tion of bone marrow– derived endothelial progenitor cells
(rosa-26, GFP, lacZ) was used to assess the incorporation of                                and, thus, may lead to a lower percentage of incorporation of
bone marrow-derived EPC into tissues. The basal incorpora-                                  bone marrow– derived progenitor cells. The efficiency of
tion rate of progenitor cells without tissue injury is extremely                            engraftment may additionally differ between distinct progen-
low.37 In ischemic tissue, the incorporation rate of genetically                            itor subpopulations (pure hematopoietic stem cells versus
labeled bone marrow– derived cells, which coexpress endo-                                   complete bone marrow cells). Indeed, therapeutic application
thelial marker proteins, differs from 0% to 90% incorpora-                                  of cells by intravenous infusion of ex vivo purified bone
tion.19,28,37– 41 Likewise, the extent of incorporation of bone                             marrow mononuclear cells or expanded endothelial progeni-
marrow– derived cells in cerebral vessels after stroke varies in                            tor cells led to a higher incorporation rate (Ϸ7% to 20%
the literature.42– 44 Whereas two studies reported positive                                 incorporation rate) as compared with the endogenously mo-
vessels with an average of 34% endothelial marker express-                                  bilized bone marrow– engrafted cells (Ϸ2%).9,47
ing bone marrow– derived cells,42,43 other groups could not                                    However, the number of incorporated cells with an endo-
detect endothelial marker expressing cells.44 High amounts                                  thelial phenotype into ischemic tissues is generally quite low.
(Ͼ50%) were predominantly detected in models of tumor                                       How can such a small number of cells increase neovascular-
angiogenesis.28,40 Some studies only detected bone marrow–                                  ization? A possible explanation might be that the efficiency
                           Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
Urbich and Dimmeler               Endothelial Progenitor Cells and Vascular Biology               347




                                                                                             Figure 2. Role of EPCs in vascular biol-
                                                                                             ogy. Injection of EPCs significantly
                                                                                             improve reendothelialization and neovas-
                                                                                             cularization after injury.




of neovascularization may not solely be attributable to the           be rapidly covered by bone marrow– derived cells deriving
incorporation of EPCs in newly formed vessels, but may also           from CD34ϩ hematopoietic stem cells in a dog model.2 In
be influenced by the release of proangiogenic factors in a            humans, the surface of ventricular assist devices was covered
paracrine manner. Indeed, the deletion of Tie-2–positive bone         by even more immature CD133-positive hematopoietic stem
marrow– derived cells through activation of a suicide gene            cells, which concomitantly express the VEGF-receptor 2.3
blocked tumor angiogenesis, although these cells are not              Additionally, Walter and coworkers demonstrated that circu-
integrated into the tumor vessels but are detected adjacent to        lating endothelial precursor cells can home to denuded parts
the vessel.41 Thus, EPCs may act similar to monocytes/mac-            of the artery after balloon injury.51 Bone marrow transplan-
rophages, which can increase arteriogenesis by providing              tation experiments revealed that bone marrow– derived cells
cytokines and growth factors. Indeed, EPCs cultivated from            can contribute to reendothelialization of grafts and denuded
different sources showed a marked expression of growth                arteries.51–53 However, in a model of transplant arteriosclero-
factors such as VEGF, HGF, and IGF-1 (C.U., unpublished               sis, bone marrow– derived cells appear to contribute only to a
data, 2004). Moreover, adherent monocytic cells, which were           minor extent to endothelial regeneration by circulating cells.18
cultivated under similar conditions, but do not express endo-         These data again indicate that there might be at least two
thelial marker proteins, also release VEGF, HGF, and                  distinct populations of circulating cells that principally are
G-CSF.14 The release of growth factors in turn may influence          capable to contribute to reendothelialization, namely mobi-
the classical process of angiogenesis, namely the proliferation       lized cells from bone marrow and non-bone marrow– derived
and migration as well as survival of mature endothelial               cells. The latter ones may arise from circulating progenitor
cells.48 However, EPCs additionally incorporated into the             cells released by non-bone marrow sources (eg, tissue resi-
newly formed vessel structures and showed endothelial                 dent stem cells) or represent vessel wall– derived endothelial
marker protein expression in vivo. In contrast, infusion of           cells.18,51–53
macrophages, which are known to release growth fac-                      A rapid regeneration of the endothelial monolayer may
tors,49,50 but were not incorporated into vessel-like struc-          prevent restenosis development by endothelial synthesis of
tures, induced only a slight increase in neovascularization           antiproliferative mediators such as nitric oxide. Indeed, en-
after ischemia, indicating— but not proving—that the ca-              hanced incorporation of ␤-galactosidase–positive, bone mar-
pacity of EPCs to physically contribute to vessel-like                row– derived cells was associated with an accelerated reen-
structures may contribute to their potent capacity to                 dothelialization and reduction of restenosis.51,52 Similar
improve neovascularization.9 Further studies will have to             results were reported by Griese et al, who demonstrated that
be designed to elucidate the contribution of physical                 infused peripheral blood monocyte– derived EPC home to
incorporation, paracrine effects and possible effects on              bioprosthetic grafts and to balloon-injured carotid arteries, the
vessel remodeling and facilitating vessel branching to                latter being associated with a significant reduction in neoin-
EPC-mediated improvement of neovascularization.                       tima deposition.54 Likewise, infusion of bone marrow– de-
                                                                      rived CD34Ϫ/CD14ϩ mononuclear cells, which are not rep-
       EPCs and Endothelial Regeneration                              resenting the population of the “classical hemangioblast,”
In the past, the regeneration of injured endothelium has been         contributed to endothelial regeneration.13 The regenerated
attributed to the migration and proliferation of neighboring          endothelium was functionally active as shown by the release
endothelial cells. More recent studies, however, indicate that        of NO,13 which is supposed to be one of the major vasculo-
additional repair mechanisms may exist to replace denuded or          protective mechanisms. Consistently, neointima development
injured arteries. Thus, implanted Dacron grafts were shown to         was significantly reduced after cell infusion.13 Whereas the
                   Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
348      Circulation Research          August 20, 2004


regeneration of the endothelium by EPCs protects lesion             apy with plasmids encoding for VEGF demonstrated an
formation, bone marrow– derived stem/progenitor cells may           augmentation of EPC levels in humans.71 Additional factors
also contribute to plaque angiogenesis, thereby potentially         inducing mobilization of progenitor cells from the bone
facilitating plaque instability.55 However, in a recent study,      marrow have been initially discovered in hematology to
no influence of bone marrow cell infusion on plaque compo-          harvest hematopoietic stem cells from the peripheral blood
sition was detected in nonischemic mice.56 An increase in           for bone marrow transplantation. For instance, granulocyte-
plaque size was only detected in the presence of ischemia,          colony stimulating factor (G-CSF), a cytokine, which is
suggesting that ischemia-induced release of growth factors          typically used for mobilization of CD34ϩ cells in patients,
predominantly accounts for this effect.56                           also increased the levels of circulating endothelial progenitor
   Overall, these studies implicate that regardless of the origin   cells. A related cytokine, the granulocyte monocyte-colony
of circulating endothelial progenitor cells, this pool of circu-    stimulating factor (GM-CSF), augments EPC levels.30 More-
lating endothelial cells may exert an important function as an      over, erythropoietin (EPO), which stimulates proliferation
endogenous repair mechanism to maintain the integrity of the        and maturation of erythroid precursors, also increased periph-
endothelial monolayer by replacing denuded parts of the             eral blood endothelial progenitor cells in mice72 and in men.73
artery (Figure 2). One can speculate that these cells may also      The correlation between EPO serum levels and the number of
regenerate the low grade endothelial damage by ongoing              CD34ϩ or CD133ϩ hematopoietic stem cells in the bone
induction of endothelial cell apoptosis induced by risk factors     marrow in patients with ischemic coronary artery disease
for coronary artery disease (see review).57 The maintenance         further supports an important role of endogenous EPO levels
of the endothelial monolayer may prevent thrombotic com-            as a physiologic determinant of EPC mobilization.72 At
plications and atherosclerotic lesion development. Although         present, it is not clear which of the mobilizing factors most
this concept has not yet been proven, several hints from            potently elevates the levels of EPCs. SDF-1 and VEGF165
recently presented data are supportive. Thus, transplantation       showed similar effects and rapidly mobilize hematopoietic
of ApoEϪ/Ϫ mice with wild-type bone marrow reduced                  stem cells and circulating endothelial precursor cells in
atherosclerotic lesion formation.58 Moreover, various risk          animal models, whereas angiopoietin-1 induced a delayed
factors for coronary artery disease, such as diabetes, hyper-       and less pronounced mobilization of endothelial and hema-
cholesterolemia, hypertension, and smoking, affect the num-         topoietic progenitors.74,75 Whereas a similar increase in white
ber and functional activity of EPCs in healthy volunteers59         blood cell counts was achieved by G-CSF application, endo-
and in patients with coronary artery disease.60 Likewise,           thelial colonies (CFU-EC) were significantly lower in G-
diabetic mice and patients were characterized by reduced            CSF– compared with VEGF- or SDF-1–treated mice. Of
functional activity of EPCs.61– 63 In addition, factors that        note, these data should be interpreted with caution, because
reduce cardiovascular risk such as statins38,51,52,64 or exer-      the responsiveness toward cytokines may vary between dif-
cise65 elevate EPC levels, which contribute to enhanced             ferent mice strains and side-by-side comparisons in humans
endothelial repair. The balance of atheroprotective and             are lacking. Moreover, the extent of increasing neutrophil and
proatherosclerotic factors, thus, may influence EPC levels          lymphocyte levels, which may provoke proinflammatory
and subsequently reendothelialization capacity.                     responses, has to be considered for a potential therapeutic
                                                                    application.
                 Mobilization of EPCs                                  First evidence for potential pharmacological modulation of
Because EPCs contribute to reendothelialization and neovas-         systemic EPC levels by atheroprotective drugs came from
cularization, increasing the number of these cells may be an        studies using HMG-CoA reductase inhibitors (statins). Statins
attractive therapeutic tool. The mobilization of stem cells in      were shown to increase the number and the functional activity
the bone marrow is determined by the local microenviron-            of EPCs in vitro,38,76 in mice,38,76 and in patients with stable
ment, the so-called “stem cell niche,” which consists of            coronary artery disease.64 The increase in EPC numbers was
fibroblasts, osteoblasts, and endothelial cells (see review).66     associated with increased bone marrow– derived cells after
Basically, mobilizing cytokines hamper the interactions be-         balloon injury and accelerated endothelial regeneration.51,52
tween stem cells and stromal cells, which finally allow stem        Although statins were shown to increase the number of stem
cells to leave the bone marrow via transendothelial migration.      cells within the bone marrow, the mechanism for enhancing
Thereby, activation of proteinases such as elastase, cathepsin      EPC numbers and function may additionally include an
G, and matrix metalloproteinases (MMPs) cleave adhesive             increase in proliferation, mobilization, and prevention of EPC
bonds on stromal cells, which interact with integrins on            senescence and apoptosis.12,38,76 Interestingly, recent studies
hematopoietic stem cells. MMP-9 was additionally shown to           additionally demonstrated that estrogen increased the levels
cleave the membrane-bound Kit ligand (mKitL) and induces            of circulating EPCs.77,78 Moreover, exercise augmented EPC
the release of soluble Kit ligand (KitL; also known as stem         levels in mice and in men.65 The molecular signaling path-
cell factor, SCF).67 Physiologically, ischemia is believed to be    ways have not been identified thus far. However, several
the predominant signal to induce mobilization of EPCs from          studies indicate that the activation of the PI3K/Akt pathway,
the bone marrow. Ischemia thereby is believed to upregulate         which has first been shown to be activated in mature
VEGF or SDF-1,68,69 which in turn are released to the               endothelial cells by statins,79 may also play an important role
circulation and induce mobilization of progenitor cells from        in statin-induced increase in EPC levels.12,76 Likewise,
the bone marrow via a MMP-9 – dependent mecha-                      VEGF, EPO, estrogen, and exercise are well known to
nism.30,46,67,70 Furthermore, clinical studies using gene ther-     augment the PI3K/Akt-pathway. Thus, these factors may
                   Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
Urbich and Dimmeler               Endothelial Progenitor Cells and Vascular Biology                 349




                                                                                             Figure 3. Mechanism of EPC homing
                                                                                             and differentiation. Recruitment and
                                                                                             incorporation of EPCs into ischemic tis-
                                                                                             sue requires a coordinated multistep
                                                                                             process including mobilization, chemoat-
                                                                                             traction, adhesion, transmigration, migra-
                                                                                             tion, tissue invasion, and in situ differen-
                                                                                             tiation. Factors that are proposed to
                                                                                             regulate the distinct steps are indicated.




share some common signaling pathways. Given that recent               cells activated by cytokines and ischemia and the transmigra-
data showed that eNOS is essential for mobilization of bone           tion of the progenitor cells through the endothelial cell
marrow– derived stem and progenitor cells,47 one may spec-            monolayer.80 Integrins are known to mediate the adhesion of
ulate that these stimuli may increase progenitor cell mobili-         various cells including hematopoietic stem cells and leuko-
zation by PI3K/Akt-dependent activation of the NO-synthase            cytes to extracellular matrix proteins and to endothelial
within the bone marrow stromal cells. Indeed, exercise and            cells.81– 83 Integrins capable of mediating cell-cell interactions
VEGF-stimulated EPC mobilization was blunted in eNOSϪ/Ϫ               are the ␤2-integrins and the ␣4␤1-integrin. ␤1-Integrins are
mice.47,65                                                            expressed by various cell types including endothelial cells
                                                                      and hematopoietic cells, whereas ␤2-integrins are found
  Mechanism of Homing and Differentiation                             preferentially on hematopoietic cells.84 Because adhesion to
Although the improvement of adult neovascularization is               endothelial cells and transmigration events are involved in the
currently under intensive investigations, the mechanism of            in vivo homing of stem cells to tissues with active angiogen-
homing and differentiation of endothelial progenitor cells is         esis,80 integrins such as the ␤2-integrins and the ␣4␤1-integrin
poorly understood. In a previous study assessing in vivo              may be involved in the homing of progenitor cells to ischemic
homing of embryonic endothelial progenitor cells derived
                                                                      tissues. Consistent with the high expression of ␤2-integrins on
from cord blood, the circulating cells arrested within tumor
                                                                      hematopoietic stem/progenitor cells, ␤2-integrins mediate ad-
microvessels, extravasated into the interstitium, and incorpo-
                                                                      hesion and transmigration of hematopoietic stem/progenitor
rated into neovessels, suggesting that adhesion and transmi-
                                                                      cells.85,86 ␤2-Integrins (CD18/CD11) are expressed on periph-
gration are involved in the recruitment of endothelial progen-
                                                                      eral blood-derived EPCs and are required for EPC-adhesion
itor cells to sites of tumor angiogenesis.80 Thus, it is
                                                                      to endothelial cells and transendothelial migration in vitro
conceivable that ex vivo expanded adult EPCs and hemato-
poietic stem/progenitor cells may engage similar pathways             (S.D., personal communication, 2004). Moreover, hemato-
for recruitment to sites of ischemia and incorporation in             poietic stem cells (Sca-1ϩ/linϪ) lacking ␤2-integrins showed
newly forming vessels. Recruitment and incorporation of               reduced homing and a lower capacity to improve neovascu-
EPCs requires a coordinated sequence of multistep adhesive            larization after ischemia (S.D., personal communication,
and signaling events including chemoattraction, adhesion,             2004). Interestingly, the homing of inflammatory cells during
and transmigration, and finally the differentiation to endothe-       pneumonia or myocardial ischemia in ␤2-integrin– deficient
lial cells (Figure 3).                                                mice is mediated by the ␣4␤1-integrin87,88 suggesting that
                                                                      deficiency of ␤2-integrins can in part be compensated by the
Adhesion and Transendothelial Migration                               ␣4␤1-integrin. Moreover, conditional deletion of the ␣4-
The initial step of homing of progenitor cells to ischemic            integrin selectively inhibited the homing of hematopoietic
tissue involves adhesion of progenitor cells to endothelial           stem/progenitor cells to the bone marrow but not to the
                   Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
350      Circulation Research           August 20, 2004


spleen,89 suggesting that the homing of progenitor cells to          TABLE 2.     Unresolved Questions
different tissues is dependent on distinct adhesion molecules.       How to define an endothelial progenitor cell?
Furthermore, in vitro studies showed that MCP-1 stimulated
                                                                       Origin of endothelial progenitor cells?
adhesion of bone marrow– derived CD34Ϫ/CD14ϩ monocytes
                                                                       Definition of subpopulations with different functional capacities?
to the endothelium was blocked by anti–␤1-integrin antibod-
ies.13 Interestingly, in this study, adhesion of CD34Ϫ/CD14ϩ         Signals for EPC homing and differentiation in vivo?
monocytes isolated from the peripheral blood to endothelial            Optimization of ex vivo culture conditions to enhance the benefit of cell
cells was less affected by MCP-1 and was not blocked by                therapy?
anti–␤1-integrin antibodies.13 Finally, the initial cell arrest of     Influence of the severity of vascular damage on the contribution of EPCs
embryonic progenitor cell homing during tumor angiogenesis             to regeneration?
was suggested to be mediated by E- and P-selectin and                Mechanisms of action?
P-selectin glycoprotein ligand-1.80 Yet, it is important to            Transdifferentiation capacity of different progenitor cells?
underscore that this study was performed with embryonic                Importance of paracrine effects?
endothelial progenitor cells. It is conceivable that different
cell types may use distinct mechanisms for homing to sites of
                                                                     interleukins, which can attract circulating progenitor cells.13
angiogenesis.
                                                                     Whereas several studies shed some light on the mechanisms
   Cell-cell contacts and transmigration events might be less
                                                                     regulating attraction of EPCs to ischemic tissue, less is known
important for the reendothelialization of denuded arteries (in
contrast to homing of progenitor cells to ischemic tissues).         with respect to migration and tissue invasion. One may
With respect to endothelial progenitor cells, studies investi-       speculate that proteases such as cathepsins or metallopro-
gated the contribution of integrins to reendothelialization,         teases may mediate the tissue invasion of EPCs.
which is mainly driven by adhesion to extracellular matrix
                                                                     Differentiation
proteins. Adhesion of EPCs to denuded vessels appears to be
                                                                     Finally, maturation of EPCs to a functional endothelial cell
mediated by vitronectin-receptors (␣v␤3- and ␣v␤5-integrins).
                                                                     may be important for functional integration in vessels. The
Thus, inhibition of ␣v␤3- and ␣v␤5-integrins with cyclic RGD
                                                                     genetic cascades that regulate differentiation in the adult
peptides blocked reendothelialization of denuded arteries in
                                                                     system are largely unknown; however, several studies deter-
vivo, suggesting that ␣v␤3- and ␣v␤5-integrins are involved in
                                                                     mined the differentiation of the common mesodermal precur-
the reendothelialization of injured carotid arteries.51 How-
                                                                     sor, the hemangioblasts, during embryonic development.
ever, other integrins such as the ␤1-integrins may also mediate
                                                                     Clearly, VEGF and its receptors play a crucial role for
adhesion of progenitor cells to extracellular matrix proteins
during reendothelialization of denuded arteries.13                   stimulating endothelial differentiation in the embryonic de-
                                                                     velopment.96 –98 Likewise, VEGF induces differentiation of
Chemotaxis, Migration, and Invasion                                  endothelial cells in ex vivo culture assays using a variety of
Given the low numbers of circulating progenitor cells, che-          adult progenitor populations (CD34ϩ,1 CD133ϩ,4 peripheral
moattraction may be of utmost importance to allow for                blood mononuclear cells).15,76 Studies with embryonic stem
recruitment of reasonable numbers of progenitor cells to the         cells further revealed that the temporal regulation of Ho-
ischemic or injured tissue. Various studies examined the             meobox (Hox) genes might play an important role. Thus, the
factors influencing hematopoietic stem cell engraftment to           orphan Hox gene termed Hex (also named Prh) is required for
the bone marrow. These factors include chemokines such as            differentiation of the hemangioblast into the definitive hema-
SDF-1,90,91 lipid mediators (sphingosine-1-phosphate),92 as          topoietic progenitors and also affected endothelial differenti-
well as factors released by heterologous cells.93 The factors        ation.99 Additionally, the serine/threonine kinase Pim-1 was
attracting circulating EPCs to the ischemic tissue may be            recently discovered as a VEGF-responsive gene, which con-
similar. Indeed, SDF-1 has been proven to stimulate recruit-         tributes to endothelial differentiation out of embryonic stem
ment of progenitor cells to the ischemic tissue.31 SDF-1             cells.100
protein levels were increased during the first days after
induction of myocardial infarction.94 Moreover, overexpres-                                        Conclusion
sion of SDF-1 augmented stem cell homing and incorporation           Taken together, infusion of different hematopoietic stem cell
into ischemic tissues.31,94 Interestingly, hematopoietic stem        populations and ex vivo expanded EPCs augmented neovas-
cells were shown to be exquisitely sensitive to SDF-1 and did        cularization of tissue after ischemia, thereby providing a
not react to G-CSF or other chemokines (eg, IL-8 and                 novel therapeutic option. However, a variety of unresolved
RANTES).91 Moreover, VEGF levels are increased during                questions remain to be answered (Table 2). The crucial
ischemia and capable to act as a chemoattractive factor to           question is how to define an endothelial progenitor cell?
EPCs.68,70,71 Interestingly, the migratory capacity of EPCs or       Overall, there is consensus that endothelial progenitor cells
bone marrow cells toward VEGF and SDF-1, respectively,               can derive from the bone marrow and that CD133/VEGFR2
determined the functional improvement of patients after stem         cells represent a population with endothelial progenitor ca-
cell therapy.95 Beside genes, which are directly upregulated         pacity. However, increasing evidence suggest that there are
by hypoxia, the invasion of immune competent cells to the            additional bone marrow– derived cell populations (eg, my-
ischemic tissue may further augment the levels of various            eloid cells) within the blood, which also can give rise to
chemokines within the ischemic tissue, such as MCP-1 or              endothelial cells. Moreover, non-bone marrow– derived cells
                    Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
Urbich and Dimmeler                      Endothelial Progenitor Cells and Vascular Biology                              351


with endothelial characteristic were isolated from the periph-                             genitor cells via regulation of cell cycle regulatory genes. Circ Res.
eral blood. This might represent shed mature endothelial cells                             2003;92:1049 –1055
                                                                                     13.   Fujiyama S, Amano K, Uehira K, Yoshida M, Nishiwaki Y, Nozawa Y,
or other endothelial cells deriving from other progenitor cell                             Jin D, Takai S, Miyazaki M, Egashira K, Imada T, Iwasaka T,
populations. Clearly, one functional assay to define endothe-                              Matsubara H. Bone marrow monocyte lineage cells adhere on injured
lial progenitor cells independent of their progeny is the                                  endothelium in a monocyte chemoattractant protein-1-dependent
                                                                                           manner and accelerate reendothelialization as endothelial progenitor
demonstration of clonal expansion activity. Possibly, func-                                cells. Circ Res. 2003;93:980 –989.
tional assays will gain additional increasing importance,                            14.   Rehman J, Li J, Orschell CM, March KL. Peripheral blood “endothelial
because recent studies suggest that endothelial progenitor                                 progenitor cells” are derived from monocyte/macrophages and secrete
cells have a favorable survival and a better response toward                               angiogenic growth factors. Circulation. 2003;107:1164 –1169.
                                                                                     15.   Kalka C, Masuda H, Takahashi T, Kalka-Moll WM, Silver M, Kearney
angiogenic growth factors compared with mature endothelial                                 M, Li T, Isner JM, Asahara T. Transplantation of ex vivo expanded
cells.101 From a therapeutic point of view, these functional                               endothelial progenitor cells for therapeutic neovascularization. Proc
activities might be more important than the source of the                                  Natl Acad Sci U S A. 2000;97:3422–3427.
                                                                                     16.   Gulati R, Jevremovic D, Peterson TE, Chatterjee S, Shah V, Vile RG,
progenitor cell. Another open question is which mechanism                                  Simari RD. Diverse origin and function of cells with endothelial phe-
underlies the improvement of neovascularization by infused                                 notype obtained from adult human blood. Circ Res. 2003;93:1023–1025.
EPCs? Likely, paracrine effects contribute in addition to the                        17.   Rookmaaker MB, Vergeer M, van Zonneveld AJ, Rabelink TJ, Verhaar
physical incorporation of EPC into newly formed capillaries.                               MC. Endothelial progenitor cells: mainly derived from the monocyte/
                                                                                           macrophage-containing CD34- mononuclear cell population and only in
The influence of the incorporation of a rather small number of                             part from the hematopoietic stem cell-containing CD34ϩ mononuclear
circulating cells on remodeling and vessel maturation has to                               cell population. Circulation. 2003;108:e150.
be further elucidated.                                                               18.   Hillebrands JL, Klatter FA, van Dijk WD, Rozing J. Bone marrow does
                                                                                           not contribute substantially to endothelial-cell replacement in transplant
                                                                                           arteriosclerosis. Nat Med. 2002;8:194 –195.
                      Acknowledgments                                                19.   Jackson KA, Majka SM, Wang H, Pocius J, Hartley CJ, Majesky MW,
This study is supported by the DFG (FOR 501: Di 600/6-1). We                               Entman ML, Michael LH, Hirschi KK, Goodell MA. Regeneration of
thank A. Aicher, E. Chavakis, C. Heeschen, and A.M. Zeiher for                             ischemic cardiac muscle and vascular endothelium by adult stem cells.
helpful discussions.                                                                       J Clin Invest. 2001;107:1395–1402.
                                                                                     20.   Reyes M, Dudek A, Jahagirdar B, Koodie L, Marker PH, Verfaillie CM.
                                                                                           Origin of endothelial progenitors in human postnatal bone marrow.
                            References                                                     J Clin Invest. 2002;109:337–346.
 1. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T,                21.   Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S,
    Witzenbichler B, Schatteman G, Isner JM. Isolation of putative pro-                    Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal-
    genitor endothelial cells for angiogenesis. Science. 1997;275:964 –967.                Ginard B, Anversa P. Adult cardiac stem cells are multipotent and
 2. Shi Q, Rafii S, Wu MH, Wijelath ES, Yu C, Ishida A, Fujita Y, Kothari                  support myocardial regeneration. Cell. 2003;114:763–776.
    S, Mohle R, Sauvage LR, Moore MA, Storb RF, Hammond WP.                          22.   Isner JM, Asahara T. Angiogenesis and vasculogenesis as therapeutic
    Evidence for circulating bone marrow-derived endothelial cells. Blood.                 strategies for postnatal neovascularization. J Clin Invest. 1999;103:
    1998;92:362–367.                                                                       1231–1236.
 3. Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, Williams M, Oz MC,              23.   Kawamoto A, Gwon HC, Iwaguro H, Yamaguchi JI, Uchida S, Masuda
    Hicklin DJ, Witte L, Moore MA, Rafii S. Expression of VEGFR-2 and                      H, Silver M, Ma H, Kearney M, Isner JM, Asahara T. Therapeutic
    AC133 by circulating human CD34(ϩ) cells identifies a population of                    potential of ex vivo expanded endothelial progenitor cells for myo-
    functional endothelial precursors. Blood. 2000;95:952–958.                             cardial ischemia. Circulation. 2001;103:634 – 637.
 4. Gehling UM, Ergun S, Schumacher U, Wagener C, Pantel K, Otte M,                  24.   Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang
    Schuch G, Schafhausen P, Mende T, Kilic N, Kluge K, Schafer B,                         J, Homma S, Edwards NM, Itescu S. Neovascularization of ischemic
    Hossfeld DK, Fiedler W. In vitro differentiation of endothelial cells                  myocardium by human bone-marrow-derived angioblasts prevents car-
    from AC133-positive progenitor cells. Blood. 2000;95:3106 –3112.                       diomyocyte apoptosis, reduces remodeling and improves cardiac
 5. Handgretinger R, Gordon PR, Leimig T, Chen X, Buhring HJ,                              function. Nat Med. 2001;7:430 – 436.
    Niethammer D, Kuci S. Biology and plasticity of CD133ϩ hemato-                   25.   Murohara T, Ikeda H, Duan J, Shintani S, Sasaki K, Eguchi H, Onitsuka
    poietic stem cells. Ann N͉Y Acad Sci. 2003;996:141–151.                                I, Matsui K, Imaizumi T. Transplanted cord blood-derived endothelial
 6. Mutin M, Canavy I, Blann A, Bory M, Sampol J, Dignat-George F.                         precursor cells augment postnatal neovascularization. J Clin Invest.
    Direct evidence of endothelial injury in acute myocardial infarction and               2000;105:1527–1536.
    unstable angina by demonstration of circulating endothelial cells. Blood.        26.   Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S,
    1999;93:2951–2958.                                                                     Masaki H, Amano K, Kishimoto Y, Yoshimoto K, Akashi H, Shimada
 7. Lin Y, Weisdorf DJ, Solovey A, Hebbel RP. Origins of circulating                       K, Iwasaka T, Imaizumi T. Therapeutic angiogenesis for patients with
    endothelial cells and endothelial outgrowth from blood. J Clin Invest.                 limb ischaemia by autologous transplantation of bone-marrow cells: a
    2000;105:71–77.                                                                        pilot study and a randomised controlled trial. Lancet. 2002;360:
 8. Schmeisser A, Garlichs CD, Zhang H, Eskafi S, Graffy C, Ludwig J,                      427– 435.
    Strasser RH, Daniel WG. Monocytes coexpress endothelial and mac-                 27.   Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert N,
    rophagocytic lineage markers and form cord-like structures in Matrigel                 Grunwald F, Aicher A, Urbich C, Martin H, Hoelzer D, Dimmeler S,
    under angiogenic conditions. Cardiovasc Res. 2001;49:671– 680.                         Zeiher AM. Transplantation of Progenitor Cells and Regeneration
 9. Urbich C, Heeschen C, Aicher A, Dernbach E, Zeiher AM, Dimmeler S.                     Enhancement in Acute Myocardial Infarction (TOPCARE-AMI). Cir-
    Relevance of monocytic features for neovascularization capacity of                     culation. 2002;106:3009 –3017.
    circulating endothelial progenitor cells. Circulation. 2003;108:                 28.   Lyden D, Hattori K, Dias S, Costa C, Blaikie P, Butros L, Chadburn A,
    2511–2516.                                                                             Heissig B, Marks W, Witte L, Wu Y, Hicklin D, Zhu Z, Hackett NR,
10. Camargo FD, Green R, Capetenaki Y, Jackson KA, Goodell MA. Single                      Crystal RG, Moore MA, Hajjar KA, Manova K, Benezra R, Rafii S.
    hematopoietic stem cells generate skeletal muscle through myeloid                      Impaired recruitment of bone-marrow-derived endothelial and hemato-
    intermediates. Nat Med. 2003;9:1520 –1527.                                             poietic precursor cells blocks tumor angiogenesis and growth. Nat Med.
11. Zhao Y, Glesne D, Huberman E. A human peripheral blood monocyte-                       2001;7:1194 –1201.
    derived subset acts as pluripotent stem cells. Proc Natl Acad Sci U S A.         29.   Schatteman GC, Hanlon HD, Jiao C, Dodds SG, Christy BA. Blood-
    2003;100:2426 –2431.                                                                   derived angioblasts accelerate blood-flow restoration in diabetic mice.
12. Assmus B, Urbich C, Aicher A, Hofmann WK, Haendeler J, Rossig L,                       J Clin Invest. 2000;106:571–578.
    Spyridopoulos I, Zeiher AM, Dimmeler S. HMG-CoA reductase inhib-                 30.   Takahashi T, Kalka C, Masuda H, Chen D, Silver M, Kearney M,
    itors reduce senescence and increase proliferation of endothelial pro-                 Magner M, Isner JM, Asahara T. Ischemia- and cytokine-induced mobi-

                      Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
352         Circulation Research                 August 20, 2004


      lization of bone marrow-derived endothelial progenitor cells for neovas-     51. Walter DH, Rittig K, Bahlmann FH, Kirchmair R, Silver M, Murayama
      cularization. Nat Med. 1999;5:434 – 438.                                         T, Nishimura H, Losordo DW, Asahara T, Isner JM. Statin therapy
31.   Yamaguchi J, Kusano KF, Masuo O, Kawamoto A, Silver M, Murasawa                  accelerates reendothelialization: a novel effect involving mobilization
      S, Bosch-Marce M, Masuda H, Losordo DW, Isner JM, Asahara T.                     and incorporation of bone marrow-derived endothelial progenitor cells.
      Stromal cell-derived factor-1 effects on ex vivo expanded endothelial            Circulation. 2002;105:3017–3024.
      progenitor cell recruitment for ischemic neovascularization. Circulation.    52. Werner N, Junk S, Laufs U, Link A, Walenta K, Bohm M, Nickenig G.
      2003;107:1322–1328.                                                              Intravenous transfusion of endothelial progenitor cells reduces neointi-
32.   Grant MB, May WS, Caballero S, Brown GA, Guthrie SM, Mames RN,                   ma formation after vascular injury. Circ Res. 2003;93:e17– e24.
      Byrne BJ, Vaught T, Spoerri PE, Peck AB, Scott EW. Adult hemato-             53. Kaushal S, Amiel GE, Guleserian KJ, Shapira OM, Perry T, Sutherland
      poietic stem cells provide functional hemangioblast activity during              FW, Rabkin E, Moran AM, Schoen FJ, Atala A, Soker S, Bischoff J,
      retinal neovascularization. Nat Med. 2002;8:607– 612.                            Mayer JE, Jr. Functional small-diameter neovessels created using endo-
33.   Hur J, Yoon CH, Kim HS, Choi JH, Kang HJ, Hwang KK, Oh BH, Lee                   thelial progenitor cells expanded ex vivo. Nat Med. 2001;7:1035–1040.
      MM, Park YB. Characterization of two types of endothelial progenitor         54. Griese DP, Ehsan A, Melo LG, Kong D, Zhang L, Mann MJ, Pratt RE,
      cells and their different contributions to neovasculogenesis. Arterioscler       Mulligan RC, Dzau VJ. Isolation and transplantation of autologous
      Thromb Vasc Biol. 2004;24:288 –293.                                              circulating endothelial cells into denuded vessels and prosthetic grafts:
34.   van Royen N, Hoefer I, Buschmann I, Kostin S, Voskuil M, Bode C,                 implications for cell-based vascular therapy. Circulation. 2003;108:
      Schaper W, Piek JJ. Effects of local MCP-1 protein therapy on the                2710 –2715.
      development of the collateral circulation and atherosclerosis in             55. Hu Y, Davison F, Zhang Z, Xu Q. Endothelial replacement and angio-
      Watanabe hyperlipidemic rabbits. Cardiovasc Res. 2003;57:178 –185.               genesis in arteriosclerotic lesions of allografts are contributed by circu-
35.   Pipp F, Heil M, Issbrucker K, Ziegelhoeffer T, Martin S, van den Heuvel          lating progenitor cells. Circulation. 2003;108:3122–3127.
      J, Weich H, Fernandez B, Golomb G, Carmeliet P, Schaper W, Clauss            56. Silvestre JS, Gojova A, Brun V, Potteaux S, Esposito B, Duriez M,
      M. VEGFR-1-selective VEGF homologue PlGF is arteriogenic:                        Clergue M, Le Ricousse-Roussanne S, Barateau V, Merval R, Groux H,
      evidence for a monocyte-mediated mechanism. Circ Res. 2003;92:                   Tobelem G, Levy B, Tedgui A, Mallat Z. Transplantation of bone
      378 –385.                                                                        marrow-derived mononuclear cells in ischemic apolipoprotein
36.   Heil M, Ziegelhoeffer T, Pipp F, Kostin S, Martin S, Clauss M, Schaper           E-knockout mice accelerates atherosclerosis without altering plaque
      W. Blood monocyte concentration is critical for enhancement of col-              composition. Circulation. 2003;108:2839 –2842.
      lateral artery growth. Am J Physiol Heart Circ Physiol. 2002;283:            57. Rossig L, Dimmeler S, Zeiher AM. Apoptosis in the vascular wall and
      H2411–H2419.                                                                     atherosclerosis. Basic Res Cardiol. 2001;96:11–22.
37.   Crosby JR, Kaminski WE, Schatteman G, Martin PJ, Raines EW, Seifert          58. Rauscher FM, Goldschmidt-Clermont PJ, Davis BH, Wang T, Gregg D,
      RA, Bowen-Pope DF. Endothelial cells of hematopoietic origin make a              Ramaswami P, Pippen AM, Annex BH, Dong C, Taylor DA. Aging,
      significant contribution to adult blood vessel formation. Circ Res. 2000;        progenitor cell exhaustion, and atherosclerosis. Circulation. 2003;108:
      87:728 –730.
                                                                                       457– 463.
38.   Llevadot J, Murasawa S, Kureishi Y, Uchida S, Masuda H, Kawamoto
                                                                                   59. Hill JM, Zalos G, Halcox JP, Schenke WH, Waclawiw MA, Quyyumi
      A, Walsh K, Isner JM, Asahara T. HMG-CoA reductase inhibitor
                                                                                       AA, Finkel T. Circulating endothelial progenitor cells, vascular
      mobilizes bone marrow– derived endothelial progenitor cells. J Clin
                                                                                       function, and cardiovascular risk. N Engl J Med. 2003;348:593– 600.
      Invest. 2001;108:399 – 405.
                                                                                   60. Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher
39.   Murayama T, Tepper OM, Silver M, Ma H, Losordo DW, Isner JM,
                                                                                       AM, Dimmeler S. Number and migratory activity of circulating endo-
      Asahara T, Kalka C. Determination of bone marrow-derived endothelial
                                                                                       thelial progenitor cells inversely correlate with risk factors for coronary
      progenitor cell significance in angiogenic growth factor-induced neo-
                                                                                       artery disease. Circ Res. 2001;89:e1– e7.
      vascularization in vivo. Exp Hematol. 2002;30:967–972.
                                                                                   61. Tepper OM, Galiano RD, Capla JM, Kalka C, Gagne PJ, Jacobowitz
40.   Garcia-Barros M, Paris F, Cordon-Cardo C, Lyden D, Rafii S,
                                                                                       GR, Levine JP, Gurtner GC. Human endothelial progenitor cells from
      Haimovitz-Friedman A, Fuks Z, Kolesnick R. Tumor response to radio-
                                                                                       type II diabetics exhibit impaired proliferation, adhesion, and incorpo-
      therapy regulated by endothelial cell apoptosis. Science. 2003;300:
                                                                                       ration into vascular structures. Circulation. 2002;106:2781–2786.
      1155–1159.
41.   De Palma M, Venneri MA, Roca C, Naldini L. Targeting exogenous               62. Loomans CJ, de Koning EJ, Staal FJ, Rookmaaker MB, Verseyden C, de
      genes to tumor angiogenesis by transplantation of genetically modified           Boer HC, Verhaar MC, Braam B, Rabelink TJ, van Zonneveld AJ.
      hematopoietic stem cells. Nat Med. 2003;9:789 –795.                              Endothelial progenitor cell dysfunction: a novel concept in the patho-
42.   Zhang ZG, Zhang L, Jiang Q, Chopp M. Bone marrow-derived endo-                   genesis of vascular complications of type 1 diabetes. Diabetes. 2004;
      thelial progenitor cells participate in cerebral neovascularization after        53:195–199.
      focal cerebral ischemia in the adult mouse. Circ Res. 2002;90:284 –288.      63. Tamarat R, Silvestre JS, Le Ricousse-Roussanne S, Barateau V,
43.   Hess DC, Hill WD, Martin-Studdard A, Carroll J, Brailer J, Carothers             Lecomte-Raclet L, Clergue M, Duriez M, Tobelem G, Levy BI.
      J. Bone marrow as a source of endothelial cells and NeuN-expressing              Impairment in ischemia-induced neovascularization in diabetes: bone
      cells After stroke. Stroke. 2002;33:1362–1368.                                   marrow mononuclear cell dysfunction and therapeutic potential of
44.   Machein MR, Renninger S, de Lima-Hahn E, Plate KH. Minor contri-                 placenta growth factor treatment. Am J Pathol. 2004;164:457– 466.
      bution of bone marrow-derived endothelial progenitors to the vascular-       64. Vasa M, Fichtlscherer S, Adler K, Aicher A, Martin H, Zeiher AM,
      ization of murine gliomas. Brain Pathol. 2003;13:582–597.                        Dimmeler S. Increase in circulating endothelial progenitor cells by statin
45.   Ziegelhoeffer T, Fernandez B, Kostin S, Heil M, Voswinckel R, Helisch            therapy in patients with stable coronary artery disease. Circulation.
      A, Schaper W. Bone marrow-derived cells do not incorporate into the              2001;103:2885–2890.
      adult growing vasculature. Circ Res. 2004;94:230 –238.                       65. Laufs U, Werner N, Link A, Endres M, Wassmann S, Jurgens K, Miche
46.   Gill M, Dias S, Hattori K, Rivera ML, Hicklin D, Witte L, Girardi L,             E, Bohm M, Nickenig G. Physical Training Increases Endothelial Pro-
      Yurt R, Himel H, Rafii S. Vascular trauma induces rapid but transient            genitor Cells, Inhibits Neointima Formation, and Enhances Angio-
      mobilization of VEGFR2(ϩ)AC133(ϩ) endothelial precursor cells. Circ              genesis. Circulation. 2004;109:220 –226.
      Res. 2001;88:167–174.                                                        66. Papayannopoulou T. Current mechanistic scenarios in hematopoietic
47.   Aicher A, Heeschen C, Mildner-Rihm C, Urbich C, Ihling C, Technau-               stem/progenitor cell mobilization. Blood. 2004;103:1580 –1585.
      Ihling K, Zeiher AM, Dimmeler S. Essential role of endothelial nitric        67. Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR, Crystal
      oxide synthase for mobilization of stem and progenitor cells. Nat Med.           RG, Besmer P, Lyden D, Moore MA, Werb Z, Rafii S. Recruitment of
      2003;9:1370 –1376.                                                               stem and progenitor cells from the bone marrow niche requires MMP-9
48.   Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other                mediated release of kit-ligand. Cell. 2002;109:625– 637.
      disease. Nat Med. 1995;1:27–31.                                              68. Lee SH, Wolf PL, Escudero R, Deutsch R, Jamieson SW, Thistlethwaite
49.   Polverini PJ, Cotran PS, Gimbrone MA Jr, Unanue ER. Activated                    PA. Early expression of angiogenesis factors in acute myocardial ische-
      macrophages induce vascular proliferation. Nature. 1977;269:804 – 806.           mia and infarction. N Engl J Med. 2000;342:626 – 633.
50.   Berse B, Brown LF, Van de Water L, Dvorak HF, Senger DR. Vascular            69. Pillarisetti K, Gupta SK. Cloning and relative expression analysis of rat
      permeability factor (vascular endothelial growth factor) gene is                 stromal cell derived factor-1 (SDF-1)1: SDF-1 alpha mRNA is selec-
      expressed differentially in normal tissues, macrophages, and tumors.             tively induced in rat model of myocardial infarction. Inflammation.
      Mol Biol Cell. 1992;3:211–220.                                                   2001;25:293–300.

                        Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
Urbich and Dimmeler                       Endothelial Progenitor Cells and Vascular Biology                            353


70. Shintani S, Murohara T, Ikeda H, Ueno T, Honma T, Katoh A, Sasaki                  86. Peled A, Grabovsky V, Habler L, Sandbank J, Arenzana-Seisdedos F,
    K, Shimada T, Oike Y, Imaizumi T. Mobilization of endothelial pro-                     Petit I, Ben-Hur H, Lapidot T, Alon R. The chemokine SDF-1 stimulates
    genitor cells in patients with acute myocardial infarction. Circulation.               integrin-mediated arrest of CD34(ϩ) cells on vascular endothelium
    2001;103:2776 –2779.                                                                   under shear flow. J Clin Invest. 1999;104:1199 –1211.
71. Kalka C, Masuda H, Takahashi T, Gordon R, Tepper O, Gravereaux E,                  87. Bowden RA, Ding ZM, Donnachie EM, Petersen TK, Michael LH,
    Pieczek A, Iwaguro H, Hayashi SI, Isner JM, Asahara T. Vascular                        Ballantyne CM, Burns AR. Role of alpha4 integrin and VCAM-1 in
    endothelial growth factor (165) gene transfer augments circulating en-                 CD18-independent neutrophil migration across mouse cardiac endothe-
    dothelial progenitor cells in human subjects. Circ Res. 2000;86:                       lium. Circ Res. 2002;90:562–569.
    1198 –1202.                                                                        88. Tasaka S, Richer SE, Mizgerd JP, Doerschuk CM. Very late antigen-4
72. Heeschen C, Aicher A, Lehmann R, Fichtlscherer S, Vasa M, Urbich C,                    in CD18-independent neutrophil emigration during acute bacterial
    Mildner-Rihm C, Martin H, Zeiher AM, Dimmeler S. Erythropoietin is                     pneumonia in mice. Am J Respir Crit Care Med. 2002;166:53– 60.
    a potent physiological stimulus for endothelial progenitor cell mobili-            89. Scott LM, Priestley GV, Papayannopoulou T. Deletion of alpha4
    zation. Blood. 2003;17:17.                                                             integrins from adult hematopoietic cells reveals roles in homeostasis,
73. Bahlmann FH, De Groot K, Spandau JM, Landry AL, Hertel B, Duckert                      regeneration, and homing. Mol Cell Biol. 2003;23:9349 –9360.
    T, Boehm SM, Menne J, Haller H, Fliser D. Erythropoietin regulates                 90. Lapidot T. Mechanism of human stem cell migration and repopulation of
    endothelial progenitor cells. Blood. 2004;103:921–926.                                 NOD/SCID and B2mnull NOD/SCID mice. The role of SDF-1/CXCR4
74. Moore MA, Hattori K, Heissig B, Shieh JH, Dias S, Crystal RG, Rafii                    interactions. Ann N Y Acad Sci. 2001;938:83–95.
    S. Mobilization of endothelial and hematopoietic stem and progenitor               91. Wright DE, Bowman EP, Wagers AJ, Butcher EC, Weissman IL. Hema-
    cells by adenovector-mediated elevation of serum levels of SDF-1,                      topoietic stem cells are uniquely selective in their migratory response to
    VEGF, and angiopoietin-1. Ann N͉Y Acad Sci. 2001;938:36 – 45; dis-                     chemokines. J Exp Med. 2002;195:1145–1154.
    cussion 45–37.                                                                     92. Kimura T, Boehmler AM, Seitz G, Kuci S, Wiesner T, Brinkmann V,
75. Hattori K, Dias S, Heissig B, Hackett NR, Lyden D, Tateno M, Hicklin                   Kanz L, Mohle R. The sphingosine 1-phosphate (S1P) receptor agonist
    DJ, Zhu Z, Witte L, Crystal RG, Moore MA, Rafii S. Vascular endo-                      FTY720 supports CXCR4-dependent migration and bone marrow
    thelial growth factor and angiopoietin-1 stimulate postnatal hemato-                   homing of human CD34ϩ progenitor cells. Blood. 2004;26:26.
    poiesis by recruitment of vasculogenic and hematopoietic stem cells. J
                                                                                       93. Adams GB, Chabner KT, Foxall RB, Weibrecht KW, Rodrigues NP,
    Exp Med. 2001;193:1005–1014.
                                                                                           Dombkowski D, Fallon R, Poznansky MC, Scadden DT. Heterologous
76. Dimmeler S, Aicher A, Vasa M, Mildner-Rihm C, Adler K, Tiemann M,
                                                                                           cells cooperate to augment stem cell migration, homing, and
    Rutten H, Fichtlscherer S, Martin H, Zeiher AM. HMG-CoA reductase
                                                                                           engraftment. Blood. 2003;101:45–51.
    inhibitors (statins) increase endothelial progenitor cells via the PI 3-ki-
                                                                                       94. Askari AT, Unzek S, Popovic ZB, Goldman CK, Forudi F, Kiedrowski
    nase/Akt pathway. J Clin Invest. 2001;108:391–397.
                                                                                           M, Rovner A, Ellis SG, Thomas JD, DiCorleto PE, Topol EJ, Penn MS.
77. Iwakura A, Luedemann C, Shastry S, Hanley A, Kearney M, Aikawa R,
                                                                                           Effect of stromal-cell-derived factor 1 on stem-cell homing and tissue
    Isner JM, Asahara T, Losordo DW. Estrogen-mediated, endothelial
                                                                                           regeneration in ischaemic cardiomyopathy. Lancet. 2003;362:697–703.
    nitric oxide synthase-dependent mobilization of bone marrow-derived
                                                                                       95. Britten MB, Abolmaali ND, Assmus B, Lehmann R, Honold J, Schmitt
    endothelial progenitor cells contributes to reendothelialization after ar-
                                                                                           J, Vogl TJ, Martin H, Schachinger V, Dimmeler S, Zeiher AM. Infarct
    terial injury. Circulation. 2003;108:3115–3121.
78. Strehlow K, Werner N, Berweiler J, Link A, Dirnagl U, Priller J, Laufs                 remodeling after intracoronary progenitor cell treatment in patients with
    K, Ghaeni L, Milosevic M, Bohm M, Nickenig G. Estrogen increases                       acute myocardial infarction (TOPCARE-AMI): mechanistic insights
    bone marrow-derived endothelial progenitor cell production and                         from serial contrast-enhanced magnetic resonance imaging. Circulation.
    diminishes neointima formation. Circulation. 2003;107:3059 –3065.                      2003;108:2212–2218.
79. Kureishi Y, Luo Z, Shiojima I, Bialik A, Fulton D, Lefer DJ, Sessa WC,             96. Ferrara N, Carver-Moore K, Chen H, Dowd M, Lu L, O’Shea KS,
    Walsh K. The HMG-CoA reductase inhibitor simvastatin activates the                     Powell-Braxton L, Hillan KJ, Moore MW. Heterozygous embryonic
    protein kinase Akt and promotes angiogenesis in normocholesterolemic                   lethality induced by targeted inactivation of the VEGF gene. Nature.
    animals. Nat Med. 2000;6:1004 –1010.                                                   1996;380:439 – 442.
80. Vajkoczy P, Blum S, Lamparter M, Mailhammer R, Erber R, Engelhardt                 97. Fong GH, Rossant J, Gertsenstein M, Breitman ML. Role of the Flt-1
    B, Vestweber D, Hatzopoulos AK. Multistep nature of microvascular                      receptor tyrosine kinase in regulating the assembly of vascular endothe-
    recruitment of ex vivo-expanded embryonic endothelial progenitor cells                 lium. Nature. 1995;376:66 –70.
    during tumor angiogenesis. J Exp Med. 2003;197:1755–1765.                          98. Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman
81. Springer TA. Traffic signals for lymphocyte recirculation and leukocyte                ML, Schuh AC. Failure of blood-island formation and vasculogenesis in
    emigration: the multistep paradigm. Cell. 1994;76:301–314.                             Flk-1-deficient mice. Nature. 1995;376:62– 66.
82. Carlos TM, Harlan JM. Leukocyte-endothelial adhesion molecules.                    99. Guo Y, Chan R, Ramsey H, Li W, Xie X, Shelley WC, Martinez-
    Blood. 1994;84:2068 –2101.                                                             Barbera JP, Bort B, Zaret K, Yoder M, Hromas R. The homeoprotein
83. Muller WA. Leukocyte-endothelial cell interactions in the inflammatory                 Hex is required for hemangioblast differentiation. Blood. 2003;102:
    response. Lab Invest. 2002;82:521–533.                                                 2428 –2435.
84. Soligo D, Schiro R, Luksch R, Manara G, Quirici N, Parravicini C,                 100. Zippo A, De Robertis A, Bardelli M, Galvagni F, Oliviero S. Identifi-
    Lambertenghi Deliliers G. Expression of integrins in human bone                        cation of Flk-1-target genes in vasculogenesis: Pim-1 is required for
    marrow. Br J Haematol. 1990;76:323–332.                                                endothelial and mural cell differentiation in vitro. Blood. 2004;24:24.
85. Kollet O, Spiegel A, Peled A, Petit I, Byk T, Hershkoviz R, Guetta E,             101. Bompais H, Chagraoui J, Canron X, Crisan M, Liu XH, Anjo A,
    Barkai G, Nagler A, Lapidot T. Rapid and efficient homing of human                     Tolla-Le Port C, Leboeuf M, Charbord P, Bikfalvi A, Uzan G. Human
    CD34(ϩ)CD38(-/low)CXCR4(ϩ) stem and progenitor cells to the bone                       endothelial cells derived from circulating progenitors display specific
    marrow and spleen of NOD/SCID and NOD/SCID/B2m(null) mice.                             functional properties as compared to mature vessel wall endothelial
    Blood. 2001;97:3283–3291.                                                              cells. Blood. 2003;20:20.




                       Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012

More Related Content

What's hot

Stem cell
Stem cellStem cell
clonogenic assay poster-2
clonogenic assay poster-2clonogenic assay poster-2
clonogenic assay poster-2
Jaclyn Brown
 
Stem cell
Stem cellStem cell
Stem cell
Manan Shah
 
Umbilical cord vs_blood_msc_source.zeddou2010_(1)
Umbilical cord vs_blood_msc_source.zeddou2010_(1)Umbilical cord vs_blood_msc_source.zeddou2010_(1)
Umbilical cord vs_blood_msc_source.zeddou2010_(1)
ComprehensiveBiologi
 
Whartons jelly review
Whartons jelly reviewWhartons jelly review
Whartons jelly review
ComprehensiveBiologi
 
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
ComprehensiveBiologi
 
Stem cell transplantation
Stem cell transplantationStem cell transplantation
Stem cell transplantation
Shilpa Reddy
 
Ch 20 -_mesenchymal_stromal_cells_from_whartons_jelly_wj-ms_cs_coupling_their...
Ch 20 -_mesenchymal_stromal_cells_from_whartons_jelly_wj-ms_cs_coupling_their...Ch 20 -_mesenchymal_stromal_cells_from_whartons_jelly_wj-ms_cs_coupling_their...
Ch 20 -_mesenchymal_stromal_cells_from_whartons_jelly_wj-ms_cs_coupling_their...
ComprehensiveBiologi
 
Cord tissuegoldstandardarticle
Cord tissuegoldstandardarticleCord tissuegoldstandardarticle
Cord tissuegoldstandardarticle
ComprehensiveBiologi
 
Exosomesfrom whartonsjelly (1)
Exosomesfrom whartonsjelly (1)Exosomesfrom whartonsjelly (1)
Exosomesfrom whartonsjelly (1)
ComprehensiveBiologi
 
Advances in stem cell technology
Advances in stem cell technologyAdvances in stem cell technology
Advances in stem cell technology
MANGLAM ARYA
 
Liver stem-cell
Liver stem-cellLiver stem-cell
Liver stem-cell
Ashraf Hassanpur
 
Haematopoietic stem cell transplantation
Haematopoietic stem cell transplantation Haematopoietic stem cell transplantation
Haematopoietic stem cell transplantation
Saber AlZahrani
 
Stem cell markers
Stem cell markersStem cell markers
Stem cell markers
Sadaf Beg
 
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applicationsUmbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
ComprehensiveBiologi
 
What is Stem Cells ? | Stem Cell Applications
What is Stem Cells ? |  Stem Cell ApplicationsWhat is Stem Cells ? |  Stem Cell Applications
What is Stem Cells ? | Stem Cell Applications
Dr Pradeep Mahajan
 
Stem cell therapy
Stem cell therapyStem cell therapy
Stem cell therapy
Karun Kumar
 
STEM CELL THERAPY
STEM CELL THERAPYSTEM CELL THERAPY
STEM CELL THERAPY
د. أنور الموسوي
 
Stem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionStem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder Dysfunction
Ankita-rastogi
 
Regenerative medicine
Regenerative medicineRegenerative medicine
Regenerative medicine
Springer
 

What's hot (20)

Stem cell
Stem cellStem cell
Stem cell
 
clonogenic assay poster-2
clonogenic assay poster-2clonogenic assay poster-2
clonogenic assay poster-2
 
Stem cell
Stem cellStem cell
Stem cell
 
Umbilical cord vs_blood_msc_source.zeddou2010_(1)
Umbilical cord vs_blood_msc_source.zeddou2010_(1)Umbilical cord vs_blood_msc_source.zeddou2010_(1)
Umbilical cord vs_blood_msc_source.zeddou2010_(1)
 
Whartons jelly review
Whartons jelly reviewWhartons jelly review
Whartons jelly review
 
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
 
Stem cell transplantation
Stem cell transplantationStem cell transplantation
Stem cell transplantation
 
Ch 20 -_mesenchymal_stromal_cells_from_whartons_jelly_wj-ms_cs_coupling_their...
Ch 20 -_mesenchymal_stromal_cells_from_whartons_jelly_wj-ms_cs_coupling_their...Ch 20 -_mesenchymal_stromal_cells_from_whartons_jelly_wj-ms_cs_coupling_their...
Ch 20 -_mesenchymal_stromal_cells_from_whartons_jelly_wj-ms_cs_coupling_their...
 
Cord tissuegoldstandardarticle
Cord tissuegoldstandardarticleCord tissuegoldstandardarticle
Cord tissuegoldstandardarticle
 
Exosomesfrom whartonsjelly (1)
Exosomesfrom whartonsjelly (1)Exosomesfrom whartonsjelly (1)
Exosomesfrom whartonsjelly (1)
 
Advances in stem cell technology
Advances in stem cell technologyAdvances in stem cell technology
Advances in stem cell technology
 
Liver stem-cell
Liver stem-cellLiver stem-cell
Liver stem-cell
 
Haematopoietic stem cell transplantation
Haematopoietic stem cell transplantation Haematopoietic stem cell transplantation
Haematopoietic stem cell transplantation
 
Stem cell markers
Stem cell markersStem cell markers
Stem cell markers
 
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applicationsUmbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
 
What is Stem Cells ? | Stem Cell Applications
What is Stem Cells ? |  Stem Cell ApplicationsWhat is Stem Cells ? |  Stem Cell Applications
What is Stem Cells ? | Stem Cell Applications
 
Stem cell therapy
Stem cell therapyStem cell therapy
Stem cell therapy
 
STEM CELL THERAPY
STEM CELL THERAPYSTEM CELL THERAPY
STEM CELL THERAPY
 
Stem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionStem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder Dysfunction
 
Regenerative medicine
Regenerative medicineRegenerative medicine
Regenerative medicine
 

Viewers also liked

Bacterias
BacteriasBacterias
Bacterias
Barbara Martinez
 
Cap 7
Cap 7Cap 7
Eye
EyeEye
Megasporogénesis/Macrosporogénesis
Megasporogénesis/MacrosporogénesisMegasporogénesis/Macrosporogénesis
Megasporogénesis/Macrosporogénesis
Barbara Martinez
 
Krebs Cycle
Krebs CycleKrebs Cycle
Krebs Cycle
scuffruff
 
Gas Exchange Surfaces
Gas Exchange SurfacesGas Exchange Surfaces
Gas Exchange Surfaces
scuffruff
 

Viewers also liked (6)

Bacterias
BacteriasBacterias
Bacterias
 
Cap 7
Cap 7Cap 7
Cap 7
 
Eye
EyeEye
Eye
 
Megasporogénesis/Macrosporogénesis
Megasporogénesis/MacrosporogénesisMegasporogénesis/Macrosporogénesis
Megasporogénesis/Macrosporogénesis
 
Krebs Cycle
Krebs CycleKrebs Cycle
Krebs Cycle
 
Gas Exchange Surfaces
Gas Exchange SurfacesGas Exchange Surfaces
Gas Exchange Surfaces
 

Similar to celulas progenitoras endoteliales Epc

STEM CELL
STEM CELLSTEM CELL
STEM CELL
PESHWA ACHARYA
 
FAQ Booklet - ReeLabs
FAQ Booklet - ReeLabsFAQ Booklet - ReeLabs
FAQ Booklet - ReeLabs
ReeLabs
 
INTRODUCTION TO STEM CELL BIOLOGY DEFINITION CLASSIFICATION AND SOURCES OF ST...
INTRODUCTION TO STEM CELL BIOLOGY DEFINITION CLASSIFICATION AND SOURCES OF ST...INTRODUCTION TO STEM CELL BIOLOGY DEFINITION CLASSIFICATION AND SOURCES OF ST...
INTRODUCTION TO STEM CELL BIOLOGY DEFINITION CLASSIFICATION AND SOURCES OF ST...
Anantha Kumar
 
hemopoiesis.ppt
hemopoiesis.ppthemopoiesis.ppt
hemopoiesis.ppt
TaniaPalChoudhury1
 
Stem-cell therapy in medicine–how far we came and what we can expect?
Stem-cell therapy in medicine–how far we came and what we can expect?Stem-cell therapy in medicine–how far we came and what we can expect?
Stem-cell therapy in medicine–how far we came and what we can expect?
Apollo Hospitals
 
Adult stem cells hope
Adult stem cells hopeAdult stem cells hope
Adult stem cells hope
bestwebsite2008
 
Adult stem cells hope
Adult stem cells hopeAdult stem cells hope
Adult stem cells hope
bestwebsite2008
 
The therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adultsThe therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adults
bestwebsite2008
 
The therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adultsThe therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adults
bestwebsite2008
 
Stem cells
Stem cellsStem cells
Stem cells
Dr.R.Dhivya.,MDS
 
Stem cells
Stem cellsStem cells
Stem cells
Vipin Shukla
 
Stem cells for Bladder Dysfunction
Stem cells for Bladder DysfunctionStem cells for Bladder Dysfunction
Stem cells for Bladder Dysfunction
Ankita-rastogi
 
Stem cells
Stem cellsStem cells
Stem cells
Islam Osman
 
Fundamentals of Stem Cells
Fundamentals of Stem CellsFundamentals of Stem Cells
Fundamentals of Stem Cells
DitoAnurogo1
 
Stem Cell
Stem CellStem Cell
Stem Cell
danggo
 
Martin Pera stem cells and the future of medicine
Martin Pera stem cells and the future of medicineMartin Pera stem cells and the future of medicine
Martin Pera stem cells and the future of medicine
igorod
 
Stem cell and regenerative medicine
Stem cell and regenerative medicineStem cell and regenerative medicine
Stem cell and regenerative medicine
Manash Paul
 
Semana 15 2010
Semana 15 2010Semana 15 2010
Semana 15 2010
Tamara Jorquiera
 
Evidence that hyperplasia is possible in the aged
Evidence that hyperplasia is possible in the agedEvidence that hyperplasia is possible in the aged
Evidence that hyperplasia is possible in the aged
Snaa Hussain
 
Name .docx
Name                                                             .docxName                                                             .docx
Name .docx
gilpinleeanna
 

Similar to celulas progenitoras endoteliales Epc (20)

STEM CELL
STEM CELLSTEM CELL
STEM CELL
 
FAQ Booklet - ReeLabs
FAQ Booklet - ReeLabsFAQ Booklet - ReeLabs
FAQ Booklet - ReeLabs
 
INTRODUCTION TO STEM CELL BIOLOGY DEFINITION CLASSIFICATION AND SOURCES OF ST...
INTRODUCTION TO STEM CELL BIOLOGY DEFINITION CLASSIFICATION AND SOURCES OF ST...INTRODUCTION TO STEM CELL BIOLOGY DEFINITION CLASSIFICATION AND SOURCES OF ST...
INTRODUCTION TO STEM CELL BIOLOGY DEFINITION CLASSIFICATION AND SOURCES OF ST...
 
hemopoiesis.ppt
hemopoiesis.ppthemopoiesis.ppt
hemopoiesis.ppt
 
Stem-cell therapy in medicine–how far we came and what we can expect?
Stem-cell therapy in medicine–how far we came and what we can expect?Stem-cell therapy in medicine–how far we came and what we can expect?
Stem-cell therapy in medicine–how far we came and what we can expect?
 
Adult stem cells hope
Adult stem cells hopeAdult stem cells hope
Adult stem cells hope
 
Adult stem cells hope
Adult stem cells hopeAdult stem cells hope
Adult stem cells hope
 
The therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adultsThe therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adults
 
The therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adultsThe therapeutic potential of stem cells from adults
The therapeutic potential of stem cells from adults
 
Stem cells
Stem cellsStem cells
Stem cells
 
Stem cells
Stem cellsStem cells
Stem cells
 
Stem cells for Bladder Dysfunction
Stem cells for Bladder DysfunctionStem cells for Bladder Dysfunction
Stem cells for Bladder Dysfunction
 
Stem cells
Stem cellsStem cells
Stem cells
 
Fundamentals of Stem Cells
Fundamentals of Stem CellsFundamentals of Stem Cells
Fundamentals of Stem Cells
 
Stem Cell
Stem CellStem Cell
Stem Cell
 
Martin Pera stem cells and the future of medicine
Martin Pera stem cells and the future of medicineMartin Pera stem cells and the future of medicine
Martin Pera stem cells and the future of medicine
 
Stem cell and regenerative medicine
Stem cell and regenerative medicineStem cell and regenerative medicine
Stem cell and regenerative medicine
 
Semana 15 2010
Semana 15 2010Semana 15 2010
Semana 15 2010
 
Evidence that hyperplasia is possible in the aged
Evidence that hyperplasia is possible in the agedEvidence that hyperplasia is possible in the aged
Evidence that hyperplasia is possible in the aged
 
Name .docx
Name                                                             .docxName                                                             .docx
Name .docx
 

celulas progenitoras endoteliales Epc

  • 1. Endothelial Progenitor Cells : Characterization and Role in Vascular Biology Carmen Urbich and Stefanie Dimmeler Circulation Research 2004, 95:343-353 doi: 10.1161/01.RES.0000137877.89448.78 Circulation Research is published by the American Heart Association. 7272 Greenville Avenue, Dallas, TX 72514 Copyright © 2004 American Heart Association. All rights reserved. Print ISSN: 0009-7330. Online ISSN: 1524-4571 The online version of this article, along with updated information and services, is located on the World Wide Web at: http://circres.ahajournals.org/content/95/4/343 Subscriptions: Information about subscribing to Circulation Research is online at http://circres.ahajournals.org//subscriptions/ Permissions: Permissions & Rights Desk, Lippincott Williams & Wilkins, a division of Wolters Kluwer Health, 351 West Camden Street, Baltimore, MD 21202-2436. Phone: 410-528-4050. Fax: 410-528-8550. E-mail: journalpermissions@lww.com Reprints: Information about reprints can be found online at http://www.lww.com/reprints Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
  • 2. Reviews This Review is part of a thematic series on Angiogenesis, which includes the following articles: Endothelial Progenitor Cells: Characterization and Role in Vascular Biology Bone Marrow–Derived Cells for Enhancing Collateral Development: Mechanisms, Animal Data, and Initial Clinical Experiences Arteriogenesis Innate Immunity and Angiogenesis Syndecans Growth Factors and Blood Vessels: Differentiation and Maturation Ralph Kelly, Guest Editor Endothelial Progenitor Cells Characterization and Role in Vascular Biology Carmen Urbich, Stefanie Dimmeler Abstract—Infusion of different hematopoietic stem cell populations and ex vivo expanded endothelial progenitor cells augments neovascularization of tissue after ischemia and contributes to reendothelialization after endothelial injury, thereby, providing a novel therapeutic option. However, controversy exists with respect to the identification and the origin of endothelial progenitor cells. Overall, there is consensus that endothelial progenitor cells can derive from the bone marrow and that CD133/VEGFR2 cells represent a population with endothelial progenitor capacity. However, increasing evidence suggests that there are additional bone marrow– derived cell populations (eg, myeloid cells, “side population” cells, and mesenchymal cells) and non-bone marrow– derived cells, which also can give rise to endothelial cells. The characterization of the different progenitor cell populations and their functional properties are discussed. Mobilization and endothelial progenitor cell–mediated neovascularization is critically regulated. Stimulatory (eg, statins and exercise) or inhibitory factors (risk factors for coronary artery disease) modulate progenitor cell levels and, thereby, affect the vascular repair capacity. Moreover, recruitment and incorporation of endothelial progenitor cells requires a coordinated sequence of multistep adhesive and signaling events including adhesion and migration (eg, by integrins), chemoattraction (eg, by SDF-1/CXCR4), and finally the differentiation to endothelial cells. This review summarizes the mechanisms regulating endothelial progenitor cell–mediated neovascularization and reendothelialization. (Circ Res. 2004;95:343-353.) Key Words: progenitor cells Ⅲ neovascularization Ⅲ vasculogenesis Ⅲ angiogenesis Ⅲ endothelial cells D ifferentiation of mesodermal cells to angioblasts and subsequent endothelial differentiation was believed to exclusively occur in embryonic development. This dogma mia. Rafii’s group in 19982 also reported the existence of “circulating bone marrow– derived endothelial progenitor cells” (CEPCs) in the adult. Again, a subset of CD34ϩ was overturned in 1997, when Asahara and colleagues1 hematopoietic stem cells was shown to differentiate to the published that purified CD34ϩ hematopoietic progenitor cells endothelial lineage and express endothelial marker proteins from adults can differentiate ex vivo to an endothelial such as vWF and incorporated Dil-Ac-LDL. Most convinc- phenotype. These cells were named “endothelial progenitor ingly, bone marrow–transplanted genetically tagged cells cells” (EPCs), showed expression of various endothelial were covering implanted Dacron grafts.2 These pioneering markers, and incorporated into neovessels at sites of ische- studies suggested the presence of circulating hemangioblasts Original received March 8, 2004; revision received May 27, 2004; accepted May 28, 2004. From Molecular Cardiology, Department of Internal Medicine IV, University of Frankfurt, Frankfurt, Germany. Correspondence to Stefanie Dimmeler, PhD, Molecular Cardiology, Dept of Internal Medicine IV, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany. E-mail Dimmeler@em.uni-frankfurt.de © 2004 American Heart Association, Inc. Circulation Research is available at http://www.circresaha.org DOI: 10.1161/01.RES.0000137877.89448.78 343 Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
  • 3. 344 Circulation Research August 20, 2004 in the adult. According to the initial discovery, EPCs or endothelial lineage. Interestingly, lineage tracking showed CEPCs were defined as cells positive for both hematopoi- that myeloid cells are the hematopoietic stem cell– derived etic stem cell markers such as CD34 and an endothelial intermediates, which contribute to muscle regeneration,10 marker protein as VEGFR2. Because CD34 is not exclu- suggesting that myeloid intermediates may be part of the sively expressed on hematopoietic stem cells but, albeit at repair capacity after injury. Moreover, a subset of human a lower level, also on mature endothelial cells, further peripheral blood monocytes acts as pluripotent stem cells.11 studies used the more immature hematopoietic stem cell Of note, a specific problem arises when cells are ex vivo marker CD1333 and demonstrated that purified CD133ϩ expanded and cultured, because the culture conditions (cul- cells can differentiate to endothelial cells in vitro.4 CD133, ture supplements such as FCS and cytokines, plastic) rapidly also known as prominin or AC133, is a highly conserved changes the phenotype of the cells. For example, supplemen- antigen with unknown biological activity, which is ex- tation of the medium with statins increased the number of pressed on hematopoietic stem cells but is absent on endothelial cell colonies isolated from mononuclear cells.12 mature endothelial cells and monocytic cells (see review).5 Moreover, continuous cultivation was shown to increase Thus, CD133ϩVEGFR2ϩ cells more likely reflect imma- endothelial marker protein expression.13 This may explain ture progenitor cells, whereas CD34ϩVEGFR2ϩ may also why different groups may obtain cells with different surface represent shedded cells of the vessel wall. At present, it is factor profile and functional activity although similar proto- unclear whether CD133 only represents a surface marker cols were used for cultivation.9,14 –16 Moreover, the interaction or has a functional activity involved in regulation of of cells within a heterogeneous mixture of cells such as the neovascularization. mononuclear cells from the blood may impact the yield and Overall, controversy exists with respect to the identifica- the functional activity of the cultivated cells.17 tion and the origin of endothelial progenitor cells, which are Generally, several studies suggested that other cell popu- isolated from peripheral blood mononuclear cells by cultiva- lations beside hematopoietic stem cells also can give rise to tion in medium favoring endothelial differentiation. In pe- endothelial cells (Figure 1). Thus, non-bone marrow– derived ripheral blood mononuclear cells, several possible sources for cells have been shown to replace the endothelial cells in endothelial cells may exist: (1) the rare number of hemato- grafts.18 In addition, adult bone marrow– derived stem/pro- poietic stem cells, (2) myeloid cells, which may differentiate genitor cells such as the side population cells and multipotent to endothelial cells under the cultivation selection pressure, adult progenitor cells, which are distinct from hematopoietic (3) other circulating progenitor cells (eg, “side population” stem cells, have also been shown to differentiate to the cells), and (4) circulating mature endothelial cells, which are endothelial lineage.19,20 Recently, tissue-resident stem cells shed off the vessel wall6 and adhere to the culture dishes. First have been isolated from the heart, which are capable to evidence that there is more than one endothelial progeny differentiate to the endothelial lineage.21 These data support within the circulating blood was provided by Hebbel and the notion that it will be difficult to define the “true” colleagues, who showed that morphological and functional endothelial progenitor cells. Overall, the field is reminiscent distinct endothelial cell populations can be grown out of to immunology, where T-cells initially were defined as one peripheral blood mononuclear cells.7 They stratified the cell population. However, the functional characterization (eg, different circulating endothelial cells according to their cytokine release and response to stimuli) helped to identify growth characteristics and morphological appearance as novel T-cell subpopulations with distinct functions and ca- “spindle-like cells,” which have a low proliferative capacity, pacities. Hopefully, better profiling of distinct cell popula- and outgrowing cells. Because the outgrowing cells showed a tions and fate mapping studies will help to identify markers, high proliferative potential and originated predominantly which distinguish the circulating endothelial precursor within from the bone marrow donors, they were considered as circu- the blood and bone marrow/non-bone marrow– derived endo- lating angioblasts.7 The authors speculated that the spindle-like thelial cells. cells may likely represent mature endothelial cells, which are shed off the vessel wall. However, this hypothesis is difficult to Role of EPCs in Neovascularization test and has not yet been proven thus far. Improvement of neovascularization is a therapeutic option to Experimentally, preplating may be a way to reduce the rescue tissue from critical ischemia.22 The finding that bone heterogeneity of the cultivated EPCs, because this excludes marrow– derived cells can home to sites of ischemia and rapidly adhering cells such as differentiated monocytic or express endothelial marker proteins has challenged the use of possible mature endothelial cells.2 However, these protocols isolated hematopoietic stem cells or EPCs for therapeutic do not eliminate myeloid and nonhematopoietic progenitor vasculogenesis. Infusion of various distinct cell types either cells, which may contribute to the ex vivo cultivated cells. isolated from the bone marrow or by ex vivo cultivation was There is increasing evidence that myeloid cells can give rise shown to augment capillary density and neovascularization of to endothelial cells as well. Specifically, CD14ϩ/CD34Ϫ ischemic tissue (Table 1 and Figure 2). In animal models of myeloid cells can coexpress endothelial markers and form myocardial infarction, the injection of ex vivo expanded tube-like structures ex vivo.8 Additionally, ex vivo expansion EPCs or stem and progenitor cells significantly improved of purified CD14ϩ mononuclear cells yielded cells with an blood flow and cardiac function and reduced left ventricular endothelial characteristic, which incorporated in newly scarring.23,24 Similarly, infusion of ex vivo expanded EPCs formed blood vessels in vivo.9 These data would suggest that deriving from peripheral blood mononuclear cells in nude myeloid cells can differentiate (or transdifferentiate) to the mice or rats improved the neovascularization in hind limb Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
  • 4. Urbich and Dimmeler Endothelial Progenitor Cells and Vascular Biology 345 Figure 1. Origin and differentiation of endothelial progenitor cells. Scheme depicts the potential origin and differenti- ation of endothelial progenitor cells from hematopoietic stem cells and nonhema- topoietic cells. ischemia models.9,15,23,25 Correspondingly, initial pilot trials uncultivated CD34Ϫ cells. Remarkably, terminally differenti- indicate that bone marrow– derived or circulating blood– ated mature endothelial cells (HMVECs, GEAECs, and derived progenitor cells are useful for therapeutically improv- SVECs) did not improve neovascularization15,24,33 suggesting ing blood supply of ischemic tissue.26,27 Autologous implan- that a not-yet-defined functional characteristic (eg, chemo- tation of bone marrow mononuclear cells in patients with kine or integrin receptors mediating homing) is essential for ischemic limbs significantly augmented ankle-brachial index EPC-mediated augmentation of blood flow after ischemia. and reduced rest pain.26 In addition, transplantation of ex vivo The functional capacity of EPCs to augment blood flow expanded endothelial progenitor cells significantly improved further does not appear to be solely attributable to a mono- coronary flow reserve and left ventricular function in patients cytic phenotype. Ex vivo cultivated EPCs from CD14ϩ with acute myocardial infarction.27 mononuclear cells or CD14Ϫ mononuclear cell starting pop- Besides models of peripheral ischemia (hind limb ische- ulation improved neovascularization to a similar extent, mia), the angiogenic potential of EPCs was also investigated whereas the same number of freshly isolated mononuclear in animal models of tumor angiogenesis. Thereby, the inhi- cells taken from the starting culture did not.9 Interestingly, bition of VEGF-responsive bone marrow– derived endothelial these experimental data are supported by first clinical trials and hematopoietic precursor cells blocks tumor angiogenesis showing that freshly isolated mononuclear cells are not well and growth.28 The use of various different models, cell suited to improve neovascularization in patients with periph- numbers, and species limits the comparability of the effi- eral vascular diseases.26 However, monocytic cells may play ciency of distinct cell populations. However, the overall a crucial role in collateral growth (arteriogenesis). Thus, the functional improvement appear similar, when isolated human attraction of monocytic cells by monocyte chemoattractant CD34ϩ, CD133ϩ, EPC, MAPC, or murine Sca-1ϩ cells were protein-1 (MCP-1) enhanced arteriogenesis.34 Moreover, de- used.4,9,15,20,23,25,29 –32 Likewise, early spindle-like cells and pletion of the monocytes reduced PlGF-induced arteriogen- late outgrowing EPCs showed comparable in vivo vasculo- esis.35 A therapeutic benefit of monocyte infusion on arterio- genic capacity.33 These results suggest that the functional genesis was demonstrated under conditions of monocyte activity of the cells to augment neovascularization is rather deficiency induced by chemical depletion.36 These data sug- independent of the type of (endothelial) progenitor cell used. gest that monocytic cells are necessary for arteriogenesis and However, the CD34Ϫ fractions of freshly isolated bone possibly neovascularization. For therapeutic application, the marrow– or blood-derived mononuclear cells showed a re- local enhancement of monocyte recruitment might be better duced incorporation and functional activity.24,29 These data suited than systemic infusion of monocytic cells, which only indicate that the number of cells capable to augment neovas- leads to a relatively minor increase in the number of circu- cularization is low in this crude fraction of freshly isolated lating monocytes. Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
  • 5. 346 Circulation Research August 20, 2004 TABLE 1. Neovascularization Induced by Injection of Progenitor Cells: Experimental and Clinical Studies Cells Surface Markers Improvement Models Incorporation Rate Experimental studies Freshly isolated cells CD34ϩ cells CD34ϩ/flk-1ϩ, CD45ϩ 1 Incorporation1 13.4 Ϯ5.7% (mouse) or 9.7 Ϯ4.5% (rabbit) Dil-Ac-LDL-EPC in CD31ϩ capillaries1 Tie-2ϩ, Dil-Ac-LDLϩ 29 Hind limb ischemia29 Frequently detected (not quantified)29 CD117bright/GATAϪ2/VEGFR2/Tie-2/AC133 24 Myocardial infarction24 20–25% of total myocardial capillary vasculature24 Sca-1ϩ BM-MNCs Sca-1ϩ 30 Hind limb ischemia30 Detected (not quantified) PBMCs T and B lymphocytes and monocytes-depleted Hind limb ischemia30 MNCs30 Ex vivo expanded cells Ex vivo expanded EPC Dil-Ac-LDLϩ/lectinϩ VEGFR2ϩ, VE-cadherinϩ, Hind limb ischemia15,31 2.1 Ϯ0.4 EPCs into vessels in a CD31ϩ, CD14ϩ, CD34ϩ 15,23 Myocardial infarction23 ϫ10 field15 241 Ϯ25 cells/mm2 (day 3) 355 Ϯ30 cells/mm2 (day 7)31 Dil-Ac-LDLϩ, NOϩ, VEGFR2ϩ, VE-cadherinϩ, Hind limb ischemia25 Frequently detected (not CD31ϩ, vWFϩ, CD45Ϫ 25 quantified)25 CD31ϩ, vWFϩ, Dil-Ac-LDLϩ, VEGFR2ϩ, Tie-2ϩ 53 Vascular graft survival, Neovessel remodeling53 80% of graft lumen at day 1553 Dil-Ac-LDLϩ/lectinϩ Hind limb ischemia9 19.8 Ϯ8% CD146ϩ/HLA-DRϩ cell VEGFR2ϩ, CD105ϩ, vWFϩ, CD45ϩ 9 containing vessels9 Early EPC: Dil-Ac-LDLϩ/lectinϩ VEGFR2ϩ, CD31ϩ, Matrigel capillaries16 ND Tie-2ϩ, VE-cadherinϪ, eNOSϪ, CD14ϩ 16 Outgrowing ECs: exhibited a greater capacity for Outgrowing ECs: VEGFR2ϩ, CD31ϩ, Tie-2ϩ, capillary morphogenesis in in vitro and in vivo VE-cadherinϩ, eNOSϩ, CD14Ϫ 16 matrigel models Early EPC: weak VEGFR1, eNOS, vWF, In vitro: late EPC showed better incorporation and Detected (not quantified)33 VE-cadherin, VEGFR2, spindle shape33 tube formation. Early EPC: higher release of Late EPC: strong VE-cadherin, VEGFR1, VEGFR2, growth factors. In vivo: comparable vasculogenic eNOS, vWF, cobblestone morphology33 potential of early and late EPC (limb perfusion, capillary density) MAPC-derived ECs Co-purified MAPC: CD34Ϫ, VE-cadherinϪ, tumor growth/angiogenesis20 MAPC-derived ECs20 AC133ϩ, Flk-1ϩ 20 35% tumor angiogenesis, 30–45% Angioblast: CD34ϩ, VE-cadherinϩ, AC133Ϫ, ϩ 20 wound healing angiogenesis, Flk-1 undifferentiated MAPCs: 12% Clinical studies BMC and monocytes (TACT-trial) CD34ϩ/Dil-Ac-LDLϩ/lectinϩ Intramuscular injection in patients with peripheral ND ischemic disease; improved blood flow26 CPC and BMC (TOPCARE-AMI) CPC: Dil-Ac-LDLϩ/lectinϩ, VEGFR2ϩ, CD31ϩ, Intracoronary infusion in patients with AMI; ND vWFϩ, CD105ϩ; BMC: CD34ϩ/CD45ϩ, increase in coronary flow reserve27 CD34ϩ/CD133ϩ, CD34ϩ/VEGFR2ϩ Mechanisms by Which EPC derived cells adjacent to vessels, which do not express Improve Neovascularization endothelial marker proteins.41,45 A reasonable explanation Although the role of EPCs in neovascularization has been might be that the model of ischemia (eg, intensity of injury or convincingly shown by several groups, the question remains: ischemia)46 significantly influences the incorporation rate. A how do EPCs improve neovascularization? minor ischemia might not as profoundly induce a mobiliza- Bone marrow transplantation of genetically modified cells tion of bone marrow– derived endothelial progenitor cells (rosa-26, GFP, lacZ) was used to assess the incorporation of and, thus, may lead to a lower percentage of incorporation of bone marrow-derived EPC into tissues. The basal incorpora- bone marrow– derived progenitor cells. The efficiency of tion rate of progenitor cells without tissue injury is extremely engraftment may additionally differ between distinct progen- low.37 In ischemic tissue, the incorporation rate of genetically itor subpopulations (pure hematopoietic stem cells versus labeled bone marrow– derived cells, which coexpress endo- complete bone marrow cells). Indeed, therapeutic application thelial marker proteins, differs from 0% to 90% incorpora- of cells by intravenous infusion of ex vivo purified bone tion.19,28,37– 41 Likewise, the extent of incorporation of bone marrow mononuclear cells or expanded endothelial progeni- marrow– derived cells in cerebral vessels after stroke varies in tor cells led to a higher incorporation rate (Ϸ7% to 20% the literature.42– 44 Whereas two studies reported positive incorporation rate) as compared with the endogenously mo- vessels with an average of 34% endothelial marker express- bilized bone marrow– engrafted cells (Ϸ2%).9,47 ing bone marrow– derived cells,42,43 other groups could not However, the number of incorporated cells with an endo- detect endothelial marker expressing cells.44 High amounts thelial phenotype into ischemic tissues is generally quite low. (Ͼ50%) were predominantly detected in models of tumor How can such a small number of cells increase neovascular- angiogenesis.28,40 Some studies only detected bone marrow– ization? A possible explanation might be that the efficiency Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
  • 6. Urbich and Dimmeler Endothelial Progenitor Cells and Vascular Biology 347 Figure 2. Role of EPCs in vascular biol- ogy. Injection of EPCs significantly improve reendothelialization and neovas- cularization after injury. of neovascularization may not solely be attributable to the be rapidly covered by bone marrow– derived cells deriving incorporation of EPCs in newly formed vessels, but may also from CD34ϩ hematopoietic stem cells in a dog model.2 In be influenced by the release of proangiogenic factors in a humans, the surface of ventricular assist devices was covered paracrine manner. Indeed, the deletion of Tie-2–positive bone by even more immature CD133-positive hematopoietic stem marrow– derived cells through activation of a suicide gene cells, which concomitantly express the VEGF-receptor 2.3 blocked tumor angiogenesis, although these cells are not Additionally, Walter and coworkers demonstrated that circu- integrated into the tumor vessels but are detected adjacent to lating endothelial precursor cells can home to denuded parts the vessel.41 Thus, EPCs may act similar to monocytes/mac- of the artery after balloon injury.51 Bone marrow transplan- rophages, which can increase arteriogenesis by providing tation experiments revealed that bone marrow– derived cells cytokines and growth factors. Indeed, EPCs cultivated from can contribute to reendothelialization of grafts and denuded different sources showed a marked expression of growth arteries.51–53 However, in a model of transplant arteriosclero- factors such as VEGF, HGF, and IGF-1 (C.U., unpublished sis, bone marrow– derived cells appear to contribute only to a data, 2004). Moreover, adherent monocytic cells, which were minor extent to endothelial regeneration by circulating cells.18 cultivated under similar conditions, but do not express endo- These data again indicate that there might be at least two thelial marker proteins, also release VEGF, HGF, and distinct populations of circulating cells that principally are G-CSF.14 The release of growth factors in turn may influence capable to contribute to reendothelialization, namely mobi- the classical process of angiogenesis, namely the proliferation lized cells from bone marrow and non-bone marrow– derived and migration as well as survival of mature endothelial cells. The latter ones may arise from circulating progenitor cells.48 However, EPCs additionally incorporated into the cells released by non-bone marrow sources (eg, tissue resi- newly formed vessel structures and showed endothelial dent stem cells) or represent vessel wall– derived endothelial marker protein expression in vivo. In contrast, infusion of cells.18,51–53 macrophages, which are known to release growth fac- A rapid regeneration of the endothelial monolayer may tors,49,50 but were not incorporated into vessel-like struc- prevent restenosis development by endothelial synthesis of tures, induced only a slight increase in neovascularization antiproliferative mediators such as nitric oxide. Indeed, en- after ischemia, indicating— but not proving—that the ca- hanced incorporation of ␤-galactosidase–positive, bone mar- pacity of EPCs to physically contribute to vessel-like row– derived cells was associated with an accelerated reen- structures may contribute to their potent capacity to dothelialization and reduction of restenosis.51,52 Similar improve neovascularization.9 Further studies will have to results were reported by Griese et al, who demonstrated that be designed to elucidate the contribution of physical infused peripheral blood monocyte– derived EPC home to incorporation, paracrine effects and possible effects on bioprosthetic grafts and to balloon-injured carotid arteries, the vessel remodeling and facilitating vessel branching to latter being associated with a significant reduction in neoin- EPC-mediated improvement of neovascularization. tima deposition.54 Likewise, infusion of bone marrow– de- rived CD34Ϫ/CD14ϩ mononuclear cells, which are not rep- EPCs and Endothelial Regeneration resenting the population of the “classical hemangioblast,” In the past, the regeneration of injured endothelium has been contributed to endothelial regeneration.13 The regenerated attributed to the migration and proliferation of neighboring endothelium was functionally active as shown by the release endothelial cells. More recent studies, however, indicate that of NO,13 which is supposed to be one of the major vasculo- additional repair mechanisms may exist to replace denuded or protective mechanisms. Consistently, neointima development injured arteries. Thus, implanted Dacron grafts were shown to was significantly reduced after cell infusion.13 Whereas the Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
  • 7. 348 Circulation Research August 20, 2004 regeneration of the endothelium by EPCs protects lesion apy with plasmids encoding for VEGF demonstrated an formation, bone marrow– derived stem/progenitor cells may augmentation of EPC levels in humans.71 Additional factors also contribute to plaque angiogenesis, thereby potentially inducing mobilization of progenitor cells from the bone facilitating plaque instability.55 However, in a recent study, marrow have been initially discovered in hematology to no influence of bone marrow cell infusion on plaque compo- harvest hematopoietic stem cells from the peripheral blood sition was detected in nonischemic mice.56 An increase in for bone marrow transplantation. For instance, granulocyte- plaque size was only detected in the presence of ischemia, colony stimulating factor (G-CSF), a cytokine, which is suggesting that ischemia-induced release of growth factors typically used for mobilization of CD34ϩ cells in patients, predominantly accounts for this effect.56 also increased the levels of circulating endothelial progenitor Overall, these studies implicate that regardless of the origin cells. A related cytokine, the granulocyte monocyte-colony of circulating endothelial progenitor cells, this pool of circu- stimulating factor (GM-CSF), augments EPC levels.30 More- lating endothelial cells may exert an important function as an over, erythropoietin (EPO), which stimulates proliferation endogenous repair mechanism to maintain the integrity of the and maturation of erythroid precursors, also increased periph- endothelial monolayer by replacing denuded parts of the eral blood endothelial progenitor cells in mice72 and in men.73 artery (Figure 2). One can speculate that these cells may also The correlation between EPO serum levels and the number of regenerate the low grade endothelial damage by ongoing CD34ϩ or CD133ϩ hematopoietic stem cells in the bone induction of endothelial cell apoptosis induced by risk factors marrow in patients with ischemic coronary artery disease for coronary artery disease (see review).57 The maintenance further supports an important role of endogenous EPO levels of the endothelial monolayer may prevent thrombotic com- as a physiologic determinant of EPC mobilization.72 At plications and atherosclerotic lesion development. Although present, it is not clear which of the mobilizing factors most this concept has not yet been proven, several hints from potently elevates the levels of EPCs. SDF-1 and VEGF165 recently presented data are supportive. Thus, transplantation showed similar effects and rapidly mobilize hematopoietic of ApoEϪ/Ϫ mice with wild-type bone marrow reduced stem cells and circulating endothelial precursor cells in atherosclerotic lesion formation.58 Moreover, various risk animal models, whereas angiopoietin-1 induced a delayed factors for coronary artery disease, such as diabetes, hyper- and less pronounced mobilization of endothelial and hema- cholesterolemia, hypertension, and smoking, affect the num- topoietic progenitors.74,75 Whereas a similar increase in white ber and functional activity of EPCs in healthy volunteers59 blood cell counts was achieved by G-CSF application, endo- and in patients with coronary artery disease.60 Likewise, thelial colonies (CFU-EC) were significantly lower in G- diabetic mice and patients were characterized by reduced CSF– compared with VEGF- or SDF-1–treated mice. Of functional activity of EPCs.61– 63 In addition, factors that note, these data should be interpreted with caution, because reduce cardiovascular risk such as statins38,51,52,64 or exer- the responsiveness toward cytokines may vary between dif- cise65 elevate EPC levels, which contribute to enhanced ferent mice strains and side-by-side comparisons in humans endothelial repair. The balance of atheroprotective and are lacking. Moreover, the extent of increasing neutrophil and proatherosclerotic factors, thus, may influence EPC levels lymphocyte levels, which may provoke proinflammatory and subsequently reendothelialization capacity. responses, has to be considered for a potential therapeutic application. Mobilization of EPCs First evidence for potential pharmacological modulation of Because EPCs contribute to reendothelialization and neovas- systemic EPC levels by atheroprotective drugs came from cularization, increasing the number of these cells may be an studies using HMG-CoA reductase inhibitors (statins). Statins attractive therapeutic tool. The mobilization of stem cells in were shown to increase the number and the functional activity the bone marrow is determined by the local microenviron- of EPCs in vitro,38,76 in mice,38,76 and in patients with stable ment, the so-called “stem cell niche,” which consists of coronary artery disease.64 The increase in EPC numbers was fibroblasts, osteoblasts, and endothelial cells (see review).66 associated with increased bone marrow– derived cells after Basically, mobilizing cytokines hamper the interactions be- balloon injury and accelerated endothelial regeneration.51,52 tween stem cells and stromal cells, which finally allow stem Although statins were shown to increase the number of stem cells to leave the bone marrow via transendothelial migration. cells within the bone marrow, the mechanism for enhancing Thereby, activation of proteinases such as elastase, cathepsin EPC numbers and function may additionally include an G, and matrix metalloproteinases (MMPs) cleave adhesive increase in proliferation, mobilization, and prevention of EPC bonds on stromal cells, which interact with integrins on senescence and apoptosis.12,38,76 Interestingly, recent studies hematopoietic stem cells. MMP-9 was additionally shown to additionally demonstrated that estrogen increased the levels cleave the membrane-bound Kit ligand (mKitL) and induces of circulating EPCs.77,78 Moreover, exercise augmented EPC the release of soluble Kit ligand (KitL; also known as stem levels in mice and in men.65 The molecular signaling path- cell factor, SCF).67 Physiologically, ischemia is believed to be ways have not been identified thus far. However, several the predominant signal to induce mobilization of EPCs from studies indicate that the activation of the PI3K/Akt pathway, the bone marrow. Ischemia thereby is believed to upregulate which has first been shown to be activated in mature VEGF or SDF-1,68,69 which in turn are released to the endothelial cells by statins,79 may also play an important role circulation and induce mobilization of progenitor cells from in statin-induced increase in EPC levels.12,76 Likewise, the bone marrow via a MMP-9 – dependent mecha- VEGF, EPO, estrogen, and exercise are well known to nism.30,46,67,70 Furthermore, clinical studies using gene ther- augment the PI3K/Akt-pathway. Thus, these factors may Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
  • 8. Urbich and Dimmeler Endothelial Progenitor Cells and Vascular Biology 349 Figure 3. Mechanism of EPC homing and differentiation. Recruitment and incorporation of EPCs into ischemic tis- sue requires a coordinated multistep process including mobilization, chemoat- traction, adhesion, transmigration, migra- tion, tissue invasion, and in situ differen- tiation. Factors that are proposed to regulate the distinct steps are indicated. share some common signaling pathways. Given that recent cells activated by cytokines and ischemia and the transmigra- data showed that eNOS is essential for mobilization of bone tion of the progenitor cells through the endothelial cell marrow– derived stem and progenitor cells,47 one may spec- monolayer.80 Integrins are known to mediate the adhesion of ulate that these stimuli may increase progenitor cell mobili- various cells including hematopoietic stem cells and leuko- zation by PI3K/Akt-dependent activation of the NO-synthase cytes to extracellular matrix proteins and to endothelial within the bone marrow stromal cells. Indeed, exercise and cells.81– 83 Integrins capable of mediating cell-cell interactions VEGF-stimulated EPC mobilization was blunted in eNOSϪ/Ϫ are the ␤2-integrins and the ␣4␤1-integrin. ␤1-Integrins are mice.47,65 expressed by various cell types including endothelial cells and hematopoietic cells, whereas ␤2-integrins are found Mechanism of Homing and Differentiation preferentially on hematopoietic cells.84 Because adhesion to Although the improvement of adult neovascularization is endothelial cells and transmigration events are involved in the currently under intensive investigations, the mechanism of in vivo homing of stem cells to tissues with active angiogen- homing and differentiation of endothelial progenitor cells is esis,80 integrins such as the ␤2-integrins and the ␣4␤1-integrin poorly understood. In a previous study assessing in vivo may be involved in the homing of progenitor cells to ischemic homing of embryonic endothelial progenitor cells derived tissues. Consistent with the high expression of ␤2-integrins on from cord blood, the circulating cells arrested within tumor hematopoietic stem/progenitor cells, ␤2-integrins mediate ad- microvessels, extravasated into the interstitium, and incorpo- hesion and transmigration of hematopoietic stem/progenitor rated into neovessels, suggesting that adhesion and transmi- cells.85,86 ␤2-Integrins (CD18/CD11) are expressed on periph- gration are involved in the recruitment of endothelial progen- eral blood-derived EPCs and are required for EPC-adhesion itor cells to sites of tumor angiogenesis.80 Thus, it is to endothelial cells and transendothelial migration in vitro conceivable that ex vivo expanded adult EPCs and hemato- poietic stem/progenitor cells may engage similar pathways (S.D., personal communication, 2004). Moreover, hemato- for recruitment to sites of ischemia and incorporation in poietic stem cells (Sca-1ϩ/linϪ) lacking ␤2-integrins showed newly forming vessels. Recruitment and incorporation of reduced homing and a lower capacity to improve neovascu- EPCs requires a coordinated sequence of multistep adhesive larization after ischemia (S.D., personal communication, and signaling events including chemoattraction, adhesion, 2004). Interestingly, the homing of inflammatory cells during and transmigration, and finally the differentiation to endothe- pneumonia or myocardial ischemia in ␤2-integrin– deficient lial cells (Figure 3). mice is mediated by the ␣4␤1-integrin87,88 suggesting that deficiency of ␤2-integrins can in part be compensated by the Adhesion and Transendothelial Migration ␣4␤1-integrin. Moreover, conditional deletion of the ␣4- The initial step of homing of progenitor cells to ischemic integrin selectively inhibited the homing of hematopoietic tissue involves adhesion of progenitor cells to endothelial stem/progenitor cells to the bone marrow but not to the Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
  • 9. 350 Circulation Research August 20, 2004 spleen,89 suggesting that the homing of progenitor cells to TABLE 2. Unresolved Questions different tissues is dependent on distinct adhesion molecules. How to define an endothelial progenitor cell? Furthermore, in vitro studies showed that MCP-1 stimulated Origin of endothelial progenitor cells? adhesion of bone marrow– derived CD34Ϫ/CD14ϩ monocytes Definition of subpopulations with different functional capacities? to the endothelium was blocked by anti–␤1-integrin antibod- ies.13 Interestingly, in this study, adhesion of CD34Ϫ/CD14ϩ Signals for EPC homing and differentiation in vivo? monocytes isolated from the peripheral blood to endothelial Optimization of ex vivo culture conditions to enhance the benefit of cell cells was less affected by MCP-1 and was not blocked by therapy? anti–␤1-integrin antibodies.13 Finally, the initial cell arrest of Influence of the severity of vascular damage on the contribution of EPCs embryonic progenitor cell homing during tumor angiogenesis to regeneration? was suggested to be mediated by E- and P-selectin and Mechanisms of action? P-selectin glycoprotein ligand-1.80 Yet, it is important to Transdifferentiation capacity of different progenitor cells? underscore that this study was performed with embryonic Importance of paracrine effects? endothelial progenitor cells. It is conceivable that different cell types may use distinct mechanisms for homing to sites of interleukins, which can attract circulating progenitor cells.13 angiogenesis. Whereas several studies shed some light on the mechanisms Cell-cell contacts and transmigration events might be less regulating attraction of EPCs to ischemic tissue, less is known important for the reendothelialization of denuded arteries (in contrast to homing of progenitor cells to ischemic tissues). with respect to migration and tissue invasion. One may With respect to endothelial progenitor cells, studies investi- speculate that proteases such as cathepsins or metallopro- gated the contribution of integrins to reendothelialization, teases may mediate the tissue invasion of EPCs. which is mainly driven by adhesion to extracellular matrix Differentiation proteins. Adhesion of EPCs to denuded vessels appears to be Finally, maturation of EPCs to a functional endothelial cell mediated by vitronectin-receptors (␣v␤3- and ␣v␤5-integrins). may be important for functional integration in vessels. The Thus, inhibition of ␣v␤3- and ␣v␤5-integrins with cyclic RGD genetic cascades that regulate differentiation in the adult peptides blocked reendothelialization of denuded arteries in system are largely unknown; however, several studies deter- vivo, suggesting that ␣v␤3- and ␣v␤5-integrins are involved in mined the differentiation of the common mesodermal precur- the reendothelialization of injured carotid arteries.51 How- sor, the hemangioblasts, during embryonic development. ever, other integrins such as the ␤1-integrins may also mediate Clearly, VEGF and its receptors play a crucial role for adhesion of progenitor cells to extracellular matrix proteins during reendothelialization of denuded arteries.13 stimulating endothelial differentiation in the embryonic de- velopment.96 –98 Likewise, VEGF induces differentiation of Chemotaxis, Migration, and Invasion endothelial cells in ex vivo culture assays using a variety of Given the low numbers of circulating progenitor cells, che- adult progenitor populations (CD34ϩ,1 CD133ϩ,4 peripheral moattraction may be of utmost importance to allow for blood mononuclear cells).15,76 Studies with embryonic stem recruitment of reasonable numbers of progenitor cells to the cells further revealed that the temporal regulation of Ho- ischemic or injured tissue. Various studies examined the meobox (Hox) genes might play an important role. Thus, the factors influencing hematopoietic stem cell engraftment to orphan Hox gene termed Hex (also named Prh) is required for the bone marrow. These factors include chemokines such as differentiation of the hemangioblast into the definitive hema- SDF-1,90,91 lipid mediators (sphingosine-1-phosphate),92 as topoietic progenitors and also affected endothelial differenti- well as factors released by heterologous cells.93 The factors ation.99 Additionally, the serine/threonine kinase Pim-1 was attracting circulating EPCs to the ischemic tissue may be recently discovered as a VEGF-responsive gene, which con- similar. Indeed, SDF-1 has been proven to stimulate recruit- tributes to endothelial differentiation out of embryonic stem ment of progenitor cells to the ischemic tissue.31 SDF-1 cells.100 protein levels were increased during the first days after induction of myocardial infarction.94 Moreover, overexpres- Conclusion sion of SDF-1 augmented stem cell homing and incorporation Taken together, infusion of different hematopoietic stem cell into ischemic tissues.31,94 Interestingly, hematopoietic stem populations and ex vivo expanded EPCs augmented neovas- cells were shown to be exquisitely sensitive to SDF-1 and did cularization of tissue after ischemia, thereby providing a not react to G-CSF or other chemokines (eg, IL-8 and novel therapeutic option. However, a variety of unresolved RANTES).91 Moreover, VEGF levels are increased during questions remain to be answered (Table 2). The crucial ischemia and capable to act as a chemoattractive factor to question is how to define an endothelial progenitor cell? EPCs.68,70,71 Interestingly, the migratory capacity of EPCs or Overall, there is consensus that endothelial progenitor cells bone marrow cells toward VEGF and SDF-1, respectively, can derive from the bone marrow and that CD133/VEGFR2 determined the functional improvement of patients after stem cells represent a population with endothelial progenitor ca- cell therapy.95 Beside genes, which are directly upregulated pacity. However, increasing evidence suggest that there are by hypoxia, the invasion of immune competent cells to the additional bone marrow– derived cell populations (eg, my- ischemic tissue may further augment the levels of various eloid cells) within the blood, which also can give rise to chemokines within the ischemic tissue, such as MCP-1 or endothelial cells. Moreover, non-bone marrow– derived cells Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
  • 10. Urbich and Dimmeler Endothelial Progenitor Cells and Vascular Biology 351 with endothelial characteristic were isolated from the periph- genitor cells via regulation of cell cycle regulatory genes. Circ Res. eral blood. This might represent shed mature endothelial cells 2003;92:1049 –1055 13. Fujiyama S, Amano K, Uehira K, Yoshida M, Nishiwaki Y, Nozawa Y, or other endothelial cells deriving from other progenitor cell Jin D, Takai S, Miyazaki M, Egashira K, Imada T, Iwasaka T, populations. Clearly, one functional assay to define endothe- Matsubara H. Bone marrow monocyte lineage cells adhere on injured lial progenitor cells independent of their progeny is the endothelium in a monocyte chemoattractant protein-1-dependent manner and accelerate reendothelialization as endothelial progenitor demonstration of clonal expansion activity. Possibly, func- cells. Circ Res. 2003;93:980 –989. tional assays will gain additional increasing importance, 14. Rehman J, Li J, Orschell CM, March KL. Peripheral blood “endothelial because recent studies suggest that endothelial progenitor progenitor cells” are derived from monocyte/macrophages and secrete cells have a favorable survival and a better response toward angiogenic growth factors. Circulation. 2003;107:1164 –1169. 15. Kalka C, Masuda H, Takahashi T, Kalka-Moll WM, Silver M, Kearney angiogenic growth factors compared with mature endothelial M, Li T, Isner JM, Asahara T. Transplantation of ex vivo expanded cells.101 From a therapeutic point of view, these functional endothelial progenitor cells for therapeutic neovascularization. Proc activities might be more important than the source of the Natl Acad Sci U S A. 2000;97:3422–3427. 16. Gulati R, Jevremovic D, Peterson TE, Chatterjee S, Shah V, Vile RG, progenitor cell. Another open question is which mechanism Simari RD. Diverse origin and function of cells with endothelial phe- underlies the improvement of neovascularization by infused notype obtained from adult human blood. Circ Res. 2003;93:1023–1025. EPCs? Likely, paracrine effects contribute in addition to the 17. Rookmaaker MB, Vergeer M, van Zonneveld AJ, Rabelink TJ, Verhaar physical incorporation of EPC into newly formed capillaries. MC. Endothelial progenitor cells: mainly derived from the monocyte/ macrophage-containing CD34- mononuclear cell population and only in The influence of the incorporation of a rather small number of part from the hematopoietic stem cell-containing CD34ϩ mononuclear circulating cells on remodeling and vessel maturation has to cell population. Circulation. 2003;108:e150. be further elucidated. 18. Hillebrands JL, Klatter FA, van Dijk WD, Rozing J. Bone marrow does not contribute substantially to endothelial-cell replacement in transplant arteriosclerosis. Nat Med. 2002;8:194 –195. Acknowledgments 19. Jackson KA, Majka SM, Wang H, Pocius J, Hartley CJ, Majesky MW, This study is supported by the DFG (FOR 501: Di 600/6-1). We Entman ML, Michael LH, Hirschi KK, Goodell MA. Regeneration of thank A. Aicher, E. Chavakis, C. Heeschen, and A.M. Zeiher for ischemic cardiac muscle and vascular endothelium by adult stem cells. helpful discussions. J Clin Invest. 2001;107:1395–1402. 20. Reyes M, Dudek A, Jahagirdar B, Koodie L, Marker PH, Verfaillie CM. Origin of endothelial progenitors in human postnatal bone marrow. References J Clin Invest. 2002;109:337–346. 1. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, 21. Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Witzenbichler B, Schatteman G, Isner JM. Isolation of putative pro- Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal- genitor endothelial cells for angiogenesis. Science. 1997;275:964 –967. Ginard B, Anversa P. Adult cardiac stem cells are multipotent and 2. Shi Q, Rafii S, Wu MH, Wijelath ES, Yu C, Ishida A, Fujita Y, Kothari support myocardial regeneration. Cell. 2003;114:763–776. S, Mohle R, Sauvage LR, Moore MA, Storb RF, Hammond WP. 22. Isner JM, Asahara T. Angiogenesis and vasculogenesis as therapeutic Evidence for circulating bone marrow-derived endothelial cells. Blood. strategies for postnatal neovascularization. J Clin Invest. 1999;103: 1998;92:362–367. 1231–1236. 3. Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, Williams M, Oz MC, 23. Kawamoto A, Gwon HC, Iwaguro H, Yamaguchi JI, Uchida S, Masuda Hicklin DJ, Witte L, Moore MA, Rafii S. Expression of VEGFR-2 and H, Silver M, Ma H, Kearney M, Isner JM, Asahara T. Therapeutic AC133 by circulating human CD34(ϩ) cells identifies a population of potential of ex vivo expanded endothelial progenitor cells for myo- functional endothelial precursors. Blood. 2000;95:952–958. cardial ischemia. Circulation. 2001;103:634 – 637. 4. Gehling UM, Ergun S, Schumacher U, Wagener C, Pantel K, Otte M, 24. Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang Schuch G, Schafhausen P, Mende T, Kilic N, Kluge K, Schafer B, J, Homma S, Edwards NM, Itescu S. Neovascularization of ischemic Hossfeld DK, Fiedler W. In vitro differentiation of endothelial cells myocardium by human bone-marrow-derived angioblasts prevents car- from AC133-positive progenitor cells. Blood. 2000;95:3106 –3112. diomyocyte apoptosis, reduces remodeling and improves cardiac 5. Handgretinger R, Gordon PR, Leimig T, Chen X, Buhring HJ, function. Nat Med. 2001;7:430 – 436. Niethammer D, Kuci S. Biology and plasticity of CD133ϩ hemato- 25. Murohara T, Ikeda H, Duan J, Shintani S, Sasaki K, Eguchi H, Onitsuka poietic stem cells. Ann N͉Y Acad Sci. 2003;996:141–151. I, Matsui K, Imaizumi T. Transplanted cord blood-derived endothelial 6. Mutin M, Canavy I, Blann A, Bory M, Sampol J, Dignat-George F. precursor cells augment postnatal neovascularization. J Clin Invest. Direct evidence of endothelial injury in acute myocardial infarction and 2000;105:1527–1536. unstable angina by demonstration of circulating endothelial cells. Blood. 26. Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S, 1999;93:2951–2958. Masaki H, Amano K, Kishimoto Y, Yoshimoto K, Akashi H, Shimada 7. Lin Y, Weisdorf DJ, Solovey A, Hebbel RP. Origins of circulating K, Iwasaka T, Imaizumi T. Therapeutic angiogenesis for patients with endothelial cells and endothelial outgrowth from blood. J Clin Invest. limb ischaemia by autologous transplantation of bone-marrow cells: a 2000;105:71–77. pilot study and a randomised controlled trial. Lancet. 2002;360: 8. Schmeisser A, Garlichs CD, Zhang H, Eskafi S, Graffy C, Ludwig J, 427– 435. Strasser RH, Daniel WG. Monocytes coexpress endothelial and mac- 27. Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert N, rophagocytic lineage markers and form cord-like structures in Matrigel Grunwald F, Aicher A, Urbich C, Martin H, Hoelzer D, Dimmeler S, under angiogenic conditions. Cardiovasc Res. 2001;49:671– 680. Zeiher AM. Transplantation of Progenitor Cells and Regeneration 9. Urbich C, Heeschen C, Aicher A, Dernbach E, Zeiher AM, Dimmeler S. Enhancement in Acute Myocardial Infarction (TOPCARE-AMI). Cir- Relevance of monocytic features for neovascularization capacity of culation. 2002;106:3009 –3017. circulating endothelial progenitor cells. Circulation. 2003;108: 28. Lyden D, Hattori K, Dias S, Costa C, Blaikie P, Butros L, Chadburn A, 2511–2516. Heissig B, Marks W, Witte L, Wu Y, Hicklin D, Zhu Z, Hackett NR, 10. Camargo FD, Green R, Capetenaki Y, Jackson KA, Goodell MA. Single Crystal RG, Moore MA, Hajjar KA, Manova K, Benezra R, Rafii S. hematopoietic stem cells generate skeletal muscle through myeloid Impaired recruitment of bone-marrow-derived endothelial and hemato- intermediates. Nat Med. 2003;9:1520 –1527. poietic precursor cells blocks tumor angiogenesis and growth. Nat Med. 11. Zhao Y, Glesne D, Huberman E. A human peripheral blood monocyte- 2001;7:1194 –1201. derived subset acts as pluripotent stem cells. Proc Natl Acad Sci U S A. 29. Schatteman GC, Hanlon HD, Jiao C, Dodds SG, Christy BA. Blood- 2003;100:2426 –2431. derived angioblasts accelerate blood-flow restoration in diabetic mice. 12. Assmus B, Urbich C, Aicher A, Hofmann WK, Haendeler J, Rossig L, J Clin Invest. 2000;106:571–578. Spyridopoulos I, Zeiher AM, Dimmeler S. HMG-CoA reductase inhib- 30. Takahashi T, Kalka C, Masuda H, Chen D, Silver M, Kearney M, itors reduce senescence and increase proliferation of endothelial pro- Magner M, Isner JM, Asahara T. Ischemia- and cytokine-induced mobi- Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
  • 11. 352 Circulation Research August 20, 2004 lization of bone marrow-derived endothelial progenitor cells for neovas- 51. Walter DH, Rittig K, Bahlmann FH, Kirchmair R, Silver M, Murayama cularization. Nat Med. 1999;5:434 – 438. T, Nishimura H, Losordo DW, Asahara T, Isner JM. Statin therapy 31. Yamaguchi J, Kusano KF, Masuo O, Kawamoto A, Silver M, Murasawa accelerates reendothelialization: a novel effect involving mobilization S, Bosch-Marce M, Masuda H, Losordo DW, Isner JM, Asahara T. and incorporation of bone marrow-derived endothelial progenitor cells. Stromal cell-derived factor-1 effects on ex vivo expanded endothelial Circulation. 2002;105:3017–3024. progenitor cell recruitment for ischemic neovascularization. Circulation. 52. Werner N, Junk S, Laufs U, Link A, Walenta K, Bohm M, Nickenig G. 2003;107:1322–1328. Intravenous transfusion of endothelial progenitor cells reduces neointi- 32. Grant MB, May WS, Caballero S, Brown GA, Guthrie SM, Mames RN, ma formation after vascular injury. Circ Res. 2003;93:e17– e24. Byrne BJ, Vaught T, Spoerri PE, Peck AB, Scott EW. Adult hemato- 53. Kaushal S, Amiel GE, Guleserian KJ, Shapira OM, Perry T, Sutherland poietic stem cells provide functional hemangioblast activity during FW, Rabkin E, Moran AM, Schoen FJ, Atala A, Soker S, Bischoff J, retinal neovascularization. Nat Med. 2002;8:607– 612. Mayer JE, Jr. Functional small-diameter neovessels created using endo- 33. Hur J, Yoon CH, Kim HS, Choi JH, Kang HJ, Hwang KK, Oh BH, Lee thelial progenitor cells expanded ex vivo. Nat Med. 2001;7:1035–1040. MM, Park YB. Characterization of two types of endothelial progenitor 54. Griese DP, Ehsan A, Melo LG, Kong D, Zhang L, Mann MJ, Pratt RE, cells and their different contributions to neovasculogenesis. Arterioscler Mulligan RC, Dzau VJ. Isolation and transplantation of autologous Thromb Vasc Biol. 2004;24:288 –293. circulating endothelial cells into denuded vessels and prosthetic grafts: 34. van Royen N, Hoefer I, Buschmann I, Kostin S, Voskuil M, Bode C, implications for cell-based vascular therapy. Circulation. 2003;108: Schaper W, Piek JJ. Effects of local MCP-1 protein therapy on the 2710 –2715. development of the collateral circulation and atherosclerosis in 55. Hu Y, Davison F, Zhang Z, Xu Q. Endothelial replacement and angio- Watanabe hyperlipidemic rabbits. Cardiovasc Res. 2003;57:178 –185. genesis in arteriosclerotic lesions of allografts are contributed by circu- 35. Pipp F, Heil M, Issbrucker K, Ziegelhoeffer T, Martin S, van den Heuvel lating progenitor cells. Circulation. 2003;108:3122–3127. J, Weich H, Fernandez B, Golomb G, Carmeliet P, Schaper W, Clauss 56. Silvestre JS, Gojova A, Brun V, Potteaux S, Esposito B, Duriez M, M. VEGFR-1-selective VEGF homologue PlGF is arteriogenic: Clergue M, Le Ricousse-Roussanne S, Barateau V, Merval R, Groux H, evidence for a monocyte-mediated mechanism. Circ Res. 2003;92: Tobelem G, Levy B, Tedgui A, Mallat Z. Transplantation of bone 378 –385. marrow-derived mononuclear cells in ischemic apolipoprotein 36. Heil M, Ziegelhoeffer T, Pipp F, Kostin S, Martin S, Clauss M, Schaper E-knockout mice accelerates atherosclerosis without altering plaque W. Blood monocyte concentration is critical for enhancement of col- composition. Circulation. 2003;108:2839 –2842. lateral artery growth. Am J Physiol Heart Circ Physiol. 2002;283: 57. Rossig L, Dimmeler S, Zeiher AM. Apoptosis in the vascular wall and H2411–H2419. atherosclerosis. Basic Res Cardiol. 2001;96:11–22. 37. Crosby JR, Kaminski WE, Schatteman G, Martin PJ, Raines EW, Seifert 58. Rauscher FM, Goldschmidt-Clermont PJ, Davis BH, Wang T, Gregg D, RA, Bowen-Pope DF. Endothelial cells of hematopoietic origin make a Ramaswami P, Pippen AM, Annex BH, Dong C, Taylor DA. Aging, significant contribution to adult blood vessel formation. Circ Res. 2000; progenitor cell exhaustion, and atherosclerosis. Circulation. 2003;108: 87:728 –730. 457– 463. 38. Llevadot J, Murasawa S, Kureishi Y, Uchida S, Masuda H, Kawamoto 59. Hill JM, Zalos G, Halcox JP, Schenke WH, Waclawiw MA, Quyyumi A, Walsh K, Isner JM, Asahara T. HMG-CoA reductase inhibitor AA, Finkel T. Circulating endothelial progenitor cells, vascular mobilizes bone marrow– derived endothelial progenitor cells. J Clin function, and cardiovascular risk. N Engl J Med. 2003;348:593– 600. Invest. 2001;108:399 – 405. 60. Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher 39. Murayama T, Tepper OM, Silver M, Ma H, Losordo DW, Isner JM, AM, Dimmeler S. Number and migratory activity of circulating endo- Asahara T, Kalka C. Determination of bone marrow-derived endothelial thelial progenitor cells inversely correlate with risk factors for coronary progenitor cell significance in angiogenic growth factor-induced neo- artery disease. Circ Res. 2001;89:e1– e7. vascularization in vivo. Exp Hematol. 2002;30:967–972. 61. Tepper OM, Galiano RD, Capla JM, Kalka C, Gagne PJ, Jacobowitz 40. Garcia-Barros M, Paris F, Cordon-Cardo C, Lyden D, Rafii S, GR, Levine JP, Gurtner GC. Human endothelial progenitor cells from Haimovitz-Friedman A, Fuks Z, Kolesnick R. Tumor response to radio- type II diabetics exhibit impaired proliferation, adhesion, and incorpo- therapy regulated by endothelial cell apoptosis. Science. 2003;300: ration into vascular structures. Circulation. 2002;106:2781–2786. 1155–1159. 41. De Palma M, Venneri MA, Roca C, Naldini L. Targeting exogenous 62. Loomans CJ, de Koning EJ, Staal FJ, Rookmaaker MB, Verseyden C, de genes to tumor angiogenesis by transplantation of genetically modified Boer HC, Verhaar MC, Braam B, Rabelink TJ, van Zonneveld AJ. hematopoietic stem cells. Nat Med. 2003;9:789 –795. Endothelial progenitor cell dysfunction: a novel concept in the patho- 42. Zhang ZG, Zhang L, Jiang Q, Chopp M. Bone marrow-derived endo- genesis of vascular complications of type 1 diabetes. Diabetes. 2004; thelial progenitor cells participate in cerebral neovascularization after 53:195–199. focal cerebral ischemia in the adult mouse. Circ Res. 2002;90:284 –288. 63. Tamarat R, Silvestre JS, Le Ricousse-Roussanne S, Barateau V, 43. Hess DC, Hill WD, Martin-Studdard A, Carroll J, Brailer J, Carothers Lecomte-Raclet L, Clergue M, Duriez M, Tobelem G, Levy BI. J. Bone marrow as a source of endothelial cells and NeuN-expressing Impairment in ischemia-induced neovascularization in diabetes: bone cells After stroke. Stroke. 2002;33:1362–1368. marrow mononuclear cell dysfunction and therapeutic potential of 44. Machein MR, Renninger S, de Lima-Hahn E, Plate KH. Minor contri- placenta growth factor treatment. Am J Pathol. 2004;164:457– 466. bution of bone marrow-derived endothelial progenitors to the vascular- 64. Vasa M, Fichtlscherer S, Adler K, Aicher A, Martin H, Zeiher AM, ization of murine gliomas. Brain Pathol. 2003;13:582–597. Dimmeler S. Increase in circulating endothelial progenitor cells by statin 45. Ziegelhoeffer T, Fernandez B, Kostin S, Heil M, Voswinckel R, Helisch therapy in patients with stable coronary artery disease. Circulation. A, Schaper W. Bone marrow-derived cells do not incorporate into the 2001;103:2885–2890. adult growing vasculature. Circ Res. 2004;94:230 –238. 65. Laufs U, Werner N, Link A, Endres M, Wassmann S, Jurgens K, Miche 46. Gill M, Dias S, Hattori K, Rivera ML, Hicklin D, Witte L, Girardi L, E, Bohm M, Nickenig G. Physical Training Increases Endothelial Pro- Yurt R, Himel H, Rafii S. Vascular trauma induces rapid but transient genitor Cells, Inhibits Neointima Formation, and Enhances Angio- mobilization of VEGFR2(ϩ)AC133(ϩ) endothelial precursor cells. Circ genesis. Circulation. 2004;109:220 –226. Res. 2001;88:167–174. 66. Papayannopoulou T. Current mechanistic scenarios in hematopoietic 47. Aicher A, Heeschen C, Mildner-Rihm C, Urbich C, Ihling C, Technau- stem/progenitor cell mobilization. Blood. 2004;103:1580 –1585. Ihling K, Zeiher AM, Dimmeler S. Essential role of endothelial nitric 67. Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR, Crystal oxide synthase for mobilization of stem and progenitor cells. Nat Med. RG, Besmer P, Lyden D, Moore MA, Werb Z, Rafii S. Recruitment of 2003;9:1370 –1376. stem and progenitor cells from the bone marrow niche requires MMP-9 48. Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other mediated release of kit-ligand. Cell. 2002;109:625– 637. disease. Nat Med. 1995;1:27–31. 68. Lee SH, Wolf PL, Escudero R, Deutsch R, Jamieson SW, Thistlethwaite 49. Polverini PJ, Cotran PS, Gimbrone MA Jr, Unanue ER. Activated PA. Early expression of angiogenesis factors in acute myocardial ische- macrophages induce vascular proliferation. Nature. 1977;269:804 – 806. mia and infarction. N Engl J Med. 2000;342:626 – 633. 50. Berse B, Brown LF, Van de Water L, Dvorak HF, Senger DR. Vascular 69. Pillarisetti K, Gupta SK. Cloning and relative expression analysis of rat permeability factor (vascular endothelial growth factor) gene is stromal cell derived factor-1 (SDF-1)1: SDF-1 alpha mRNA is selec- expressed differentially in normal tissues, macrophages, and tumors. tively induced in rat model of myocardial infarction. Inflammation. Mol Biol Cell. 1992;3:211–220. 2001;25:293–300. Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012
  • 12. Urbich and Dimmeler Endothelial Progenitor Cells and Vascular Biology 353 70. Shintani S, Murohara T, Ikeda H, Ueno T, Honma T, Katoh A, Sasaki 86. Peled A, Grabovsky V, Habler L, Sandbank J, Arenzana-Seisdedos F, K, Shimada T, Oike Y, Imaizumi T. Mobilization of endothelial pro- Petit I, Ben-Hur H, Lapidot T, Alon R. The chemokine SDF-1 stimulates genitor cells in patients with acute myocardial infarction. Circulation. integrin-mediated arrest of CD34(ϩ) cells on vascular endothelium 2001;103:2776 –2779. under shear flow. J Clin Invest. 1999;104:1199 –1211. 71. Kalka C, Masuda H, Takahashi T, Gordon R, Tepper O, Gravereaux E, 87. Bowden RA, Ding ZM, Donnachie EM, Petersen TK, Michael LH, Pieczek A, Iwaguro H, Hayashi SI, Isner JM, Asahara T. Vascular Ballantyne CM, Burns AR. Role of alpha4 integrin and VCAM-1 in endothelial growth factor (165) gene transfer augments circulating en- CD18-independent neutrophil migration across mouse cardiac endothe- dothelial progenitor cells in human subjects. Circ Res. 2000;86: lium. Circ Res. 2002;90:562–569. 1198 –1202. 88. Tasaka S, Richer SE, Mizgerd JP, Doerschuk CM. Very late antigen-4 72. Heeschen C, Aicher A, Lehmann R, Fichtlscherer S, Vasa M, Urbich C, in CD18-independent neutrophil emigration during acute bacterial Mildner-Rihm C, Martin H, Zeiher AM, Dimmeler S. Erythropoietin is pneumonia in mice. Am J Respir Crit Care Med. 2002;166:53– 60. a potent physiological stimulus for endothelial progenitor cell mobili- 89. Scott LM, Priestley GV, Papayannopoulou T. Deletion of alpha4 zation. Blood. 2003;17:17. integrins from adult hematopoietic cells reveals roles in homeostasis, 73. Bahlmann FH, De Groot K, Spandau JM, Landry AL, Hertel B, Duckert regeneration, and homing. Mol Cell Biol. 2003;23:9349 –9360. T, Boehm SM, Menne J, Haller H, Fliser D. Erythropoietin regulates 90. Lapidot T. Mechanism of human stem cell migration and repopulation of endothelial progenitor cells. Blood. 2004;103:921–926. NOD/SCID and B2mnull NOD/SCID mice. The role of SDF-1/CXCR4 74. Moore MA, Hattori K, Heissig B, Shieh JH, Dias S, Crystal RG, Rafii interactions. Ann N Y Acad Sci. 2001;938:83–95. S. Mobilization of endothelial and hematopoietic stem and progenitor 91. Wright DE, Bowman EP, Wagers AJ, Butcher EC, Weissman IL. Hema- cells by adenovector-mediated elevation of serum levels of SDF-1, topoietic stem cells are uniquely selective in their migratory response to VEGF, and angiopoietin-1. Ann N͉Y Acad Sci. 2001;938:36 – 45; dis- chemokines. J Exp Med. 2002;195:1145–1154. cussion 45–37. 92. Kimura T, Boehmler AM, Seitz G, Kuci S, Wiesner T, Brinkmann V, 75. Hattori K, Dias S, Heissig B, Hackett NR, Lyden D, Tateno M, Hicklin Kanz L, Mohle R. The sphingosine 1-phosphate (S1P) receptor agonist DJ, Zhu Z, Witte L, Crystal RG, Moore MA, Rafii S. Vascular endo- FTY720 supports CXCR4-dependent migration and bone marrow thelial growth factor and angiopoietin-1 stimulate postnatal hemato- homing of human CD34ϩ progenitor cells. Blood. 2004;26:26. poiesis by recruitment of vasculogenic and hematopoietic stem cells. J 93. Adams GB, Chabner KT, Foxall RB, Weibrecht KW, Rodrigues NP, Exp Med. 2001;193:1005–1014. Dombkowski D, Fallon R, Poznansky MC, Scadden DT. Heterologous 76. Dimmeler S, Aicher A, Vasa M, Mildner-Rihm C, Adler K, Tiemann M, cells cooperate to augment stem cell migration, homing, and Rutten H, Fichtlscherer S, Martin H, Zeiher AM. HMG-CoA reductase engraftment. Blood. 2003;101:45–51. inhibitors (statins) increase endothelial progenitor cells via the PI 3-ki- 94. Askari AT, Unzek S, Popovic ZB, Goldman CK, Forudi F, Kiedrowski nase/Akt pathway. J Clin Invest. 2001;108:391–397. M, Rovner A, Ellis SG, Thomas JD, DiCorleto PE, Topol EJ, Penn MS. 77. Iwakura A, Luedemann C, Shastry S, Hanley A, Kearney M, Aikawa R, Effect of stromal-cell-derived factor 1 on stem-cell homing and tissue Isner JM, Asahara T, Losordo DW. Estrogen-mediated, endothelial regeneration in ischaemic cardiomyopathy. Lancet. 2003;362:697–703. nitric oxide synthase-dependent mobilization of bone marrow-derived 95. Britten MB, Abolmaali ND, Assmus B, Lehmann R, Honold J, Schmitt endothelial progenitor cells contributes to reendothelialization after ar- J, Vogl TJ, Martin H, Schachinger V, Dimmeler S, Zeiher AM. Infarct terial injury. Circulation. 2003;108:3115–3121. 78. Strehlow K, Werner N, Berweiler J, Link A, Dirnagl U, Priller J, Laufs remodeling after intracoronary progenitor cell treatment in patients with K, Ghaeni L, Milosevic M, Bohm M, Nickenig G. Estrogen increases acute myocardial infarction (TOPCARE-AMI): mechanistic insights bone marrow-derived endothelial progenitor cell production and from serial contrast-enhanced magnetic resonance imaging. Circulation. diminishes neointima formation. Circulation. 2003;107:3059 –3065. 2003;108:2212–2218. 79. Kureishi Y, Luo Z, Shiojima I, Bialik A, Fulton D, Lefer DJ, Sessa WC, 96. Ferrara N, Carver-Moore K, Chen H, Dowd M, Lu L, O’Shea KS, Walsh K. The HMG-CoA reductase inhibitor simvastatin activates the Powell-Braxton L, Hillan KJ, Moore MW. Heterozygous embryonic protein kinase Akt and promotes angiogenesis in normocholesterolemic lethality induced by targeted inactivation of the VEGF gene. Nature. animals. Nat Med. 2000;6:1004 –1010. 1996;380:439 – 442. 80. Vajkoczy P, Blum S, Lamparter M, Mailhammer R, Erber R, Engelhardt 97. Fong GH, Rossant J, Gertsenstein M, Breitman ML. Role of the Flt-1 B, Vestweber D, Hatzopoulos AK. Multistep nature of microvascular receptor tyrosine kinase in regulating the assembly of vascular endothe- recruitment of ex vivo-expanded embryonic endothelial progenitor cells lium. Nature. 1995;376:66 –70. during tumor angiogenesis. J Exp Med. 2003;197:1755–1765. 98. Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman 81. Springer TA. Traffic signals for lymphocyte recirculation and leukocyte ML, Schuh AC. Failure of blood-island formation and vasculogenesis in emigration: the multistep paradigm. Cell. 1994;76:301–314. Flk-1-deficient mice. Nature. 1995;376:62– 66. 82. Carlos TM, Harlan JM. Leukocyte-endothelial adhesion molecules. 99. Guo Y, Chan R, Ramsey H, Li W, Xie X, Shelley WC, Martinez- Blood. 1994;84:2068 –2101. Barbera JP, Bort B, Zaret K, Yoder M, Hromas R. The homeoprotein 83. Muller WA. Leukocyte-endothelial cell interactions in the inflammatory Hex is required for hemangioblast differentiation. Blood. 2003;102: response. Lab Invest. 2002;82:521–533. 2428 –2435. 84. Soligo D, Schiro R, Luksch R, Manara G, Quirici N, Parravicini C, 100. Zippo A, De Robertis A, Bardelli M, Galvagni F, Oliviero S. Identifi- Lambertenghi Deliliers G. Expression of integrins in human bone cation of Flk-1-target genes in vasculogenesis: Pim-1 is required for marrow. Br J Haematol. 1990;76:323–332. endothelial and mural cell differentiation in vitro. Blood. 2004;24:24. 85. Kollet O, Spiegel A, Peled A, Petit I, Byk T, Hershkoviz R, Guetta E, 101. Bompais H, Chagraoui J, Canron X, Crisan M, Liu XH, Anjo A, Barkai G, Nagler A, Lapidot T. Rapid and efficient homing of human Tolla-Le Port C, Leboeuf M, Charbord P, Bikfalvi A, Uzan G. Human CD34(ϩ)CD38(-/low)CXCR4(ϩ) stem and progenitor cells to the bone endothelial cells derived from circulating progenitors display specific marrow and spleen of NOD/SCID and NOD/SCID/B2m(null) mice. functional properties as compared to mature vessel wall endothelial Blood. 2001;97:3283–3291. cells. Blood. 2003;20:20. Downloaded from http://circres.ahajournals.org/ at UNIV DE CONCEPCION on April 11, 2012