Endothelial progenitor cells in vascular repair and remodeling

4

Click here to load reader

Transcript of Endothelial progenitor cells in vascular repair and remodeling

Page 1: Endothelial progenitor cells in vascular repair and remodeling

E

MI

a

A

KSAAC

1

bpaeaHlpceudadctef

D

1d

Pharmacological Research 58 (2008) 148–151

Contents lists available at ScienceDirect

Pharmacological Research

journa l homepage: www.e lsev ier .com/ locate /yphrs

ndothelial progenitor cells in vascular repair and remodeling

ihail Hristov, Christian Weber ∗

nstitute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Germany

r t i c l e i n f o

rticle history:Accepted 26 July 2008

eywords:tem cellsngiogenesisrterial injuryoronary artery disease

a b s t r a c t

Postnatal bone marrow contains a subtype of unique progenitor cells that have the capacity to differen-tiate into functional endothelial cells. Hence, these cells have been termed endothelial progenitor cells(EPCs). In general, circulating EPCs were characterized by the expression of CD133, CD34 and the vascularendothelial growth factor receptor-2 (VEGFR2). Recent data have additionally described some CD14+/low

myeloid subsets as functional endothelial precursors. Convincing evidence in vivo has further emergedthat the vascular homing of EPCs contributes to endothelial regeneration thereby limiting neointimalhyperplasia after arterial injury. However, in the context of primary atherosclerosis, plaque progression

and destabilization, injection of EPCs as well as application of stem-cell mobilizing factors have beenshown to correlate with conversion to unstable plaque phenotype. Clinically, the number and function ofEPCs have been positively linked with an improved endothelial function or regeneration but frequentlyinversely correlated with cardiovascular risk (factors). Thus, considering the dual contribution of EPCs invascular repair and remodeling in primary atherosclerosis versus arterial injury and identifying mech-anisms for selective control of their recruitment appears crucial to improve prediction and to directly

cular

ercalpioeftmflst

modulate endogenous vas

. Introduction

The endothelial monolayer represents a dynamic physiologicalorder between circulating blood and the surrounding tissue thatrovides a non-adhesive surface for platelets and leukocytes butlso produces a variety of important vasoregulatory factors such asndothelin, prostaglandins and nitric oxide [1]. In healthy subjects,low basal level of endothelial turnover has been described [1,2].owever, acute injury or chronic immuno-inflammatory endothe-

ial dysfunction leads to the loss of anti-thrombotic propertiesarallel to enhanced arrest and transmigration of circulating leuko-ytes. This pathological vessel remodeling gradually results inxcessive sub-endothelial accumulation of lipids and immune cells,ncontrolled proliferation of smooth muscle cells (SMCs), matrixeposition and foam cell formation [3]. Consequently, occlusivetherosclerotic plaques with luminal narrowing of the arterial wallevelop and clinically result in chronic distal tissue ischemia often

omplicated by acute stroke or myocardial infarction [3,4]. Beyondhe vascular complications above, an adequate endothelial regen-ration is also crucial for diminishing arterial stenosis secondaryollowing injury (balloon angioplasty or stent placement). This

∗ Corresponding author at: IMCAR, University Hospital Aachen, Pauwelsstr. 30,-52074 Aachen, Germany. Tel.: +49 241 80 80580; fax: +49 241 80 82716.

E-mail address: [email protected] (C. Weber).

2

iiadas

043-6618/$ – see front matter © 2008 Elsevier Ltd. All rights reserved.oi:10.1016/j.phrs.2008.07.008

homeostasis.© 2008 Elsevier Ltd. All rights reserved.

ndothelial repair can occur by migration and proliferation of sur-ounding mature endothelial cells. However, mature endothelialells are terminally differentiated with a low proliferative potentialnd their capacity to substitute damaged endothelium is altogetherimited. Therefore, the endothelial regeneration may need the sup-ort of other cell types. Accumulating evidence in the past decade

ndicates that adult peripheral blood contains a unique subtypef bone marrow-derived cells with properties similar to those ofmbryonic angioblasts [5–7]. These cells have the potential to dif-erentiate into mature endothelial cells and have therefore beenermed endothelial progenitor cells (EPCs). Recent studies in ani-

als and humans unveiled the ability of EPCs to ameliorate theunction of ischemic organs possibly by both induction and modu-ation of angiogenesis (incorporation and paracrine action) and toupport the re-endothelialization of injured arteries by replacinghe dysfunctional endothelial cells [8,9].

. Origin and characterization of EPCs

EPCs represent the most widely studied adult human progen-tor cell subpopulation up to now. These cells can be localized

n the bone marrow and peripheral blood or can reside in therterial wall [5,6,10]. Despite impressive amount of publishedata, the exact definition of EPCs remains rather controversialnd not yet consistent. This is related to the fact that differentubsets (CD34+/− and CD14+) of circulating cells were reported
Page 2: Endothelial progenitor cells in vascular repair and remodeling

ologica

tcccmat[

cmtdbcltgpcv

lem2cefeav

3m

itetTctmctobiapetTho

rtabAd

itmcrtCvtpscpFCn[pEpa

mictti[wneedtmsCs[aao

4

dafimsirlca[

M. Hristov, C. Weber / Pharmac

o acquire endothelial phenotype [5,6,11,12]. Nevertheless, markerombinations most commonly used for identifying the putative cir-ulating EPC comprise CD133+CD34+VEGFR2+ or CD34+VEGFR2+

ell subsets [5,6]. Recent reports have highlighted also circulatingyeloid subpopulations (CD14+CD34low, CD14+VEGFR2+CXCR2+/−

nd CD14lowCD16+Tie-2+) as functional angiogenic cells with con-ributions to endothelial repair and ischemic or tumor angiogenesis12–14].

EPCs were cultured by plating of separated mononuclearells on fibronectin-coated dishes in endothelial-specific growthedium. This in vitro approach results in obtaining of two cell

ypes. First, adherent spindle-shaped cells develop after 4–7ays described as “early” endothelial outgrowth from peripherallood [8,14–16]. Notably, these “early” endothelial-like cells shareommon morphological properties of monocytes and endothe-ial cells (expression of CD11b, CD11c, CD14, CD31, VE-cadherinogether with LDL-uptake and lectin-binding) and secret angio-enic cytokines [14–16]. Second, after 3–4 weeks in culture a “late”roliferative outgrowth with characteristics of mature endothelialells arises and these cells possess the ability to form functionalessels and to attenuate neointimal hyperplasia in vivo [17–20].

Accordingly, EPCs correspond to a rather heterogeneous popu-ation of multiple origins and phenotype. Their common featuresncompass expression of progenitor, myeloid and endothelialarkers (CD133, CD34, vascular endothelial growth factor receptor-(VEGFR2), CD14, CD31, VE-cadherin, etc.), clonal expansion

apacity, proliferative potential and differentiation towards thendothelial lineage. Moreover, cultured endothelial outgrowthrom bone marrow or peripheral blood could be successfullymployed for molecular analysis and functional assessment in vitros well as for transplantation in terms of pro-angiogenic therapy inivo.

. Physiological and pathological complexity of EPCobilization and homing

Under steady-state conditions, progenitor cells were maintainednactive and contact the bone marrow stroma. During mobiliza-ion, the major pool of bone marrow progenitor cells, including forxample the c-kit+ population, becomes activated and shifts fromhe quiescent stromal niche into the bone marrow sinusoids [21,22].his process is initiated by matrix metalloproteinase-9-dependentleavage of membrane bound c-kit and undergoes activation by dis-inct peripheral signals [22]. One of the most essential triggers for

obilization of stem/progenitor cells is the CXC chemokine stromalell-derived factor-1� (SDF-1�/CXCL12), which specifically binds tohe CXC chemokine receptor-4 (CXCR4) [21,23]. CXCL12 is expressedr surface-immobilized not only at the bone marrow stromal cellsut also at endothelial cells, injured SMCs and activated platelets

n the periphery [21,23]. Clinically, CXCL12 has been describeds homeostatic and anti-inflammatory chemokine with reducedlasma levels being correlated with unstable coronary artery dis-ase [24]. The gene expression of CXCL12 is mainly regulated by theranscription factor hypoxia-inducible factor-1� (HIF-1�) [21,23].he expression of HIF-1� is up regulated in injured arteries and inypoxic tissue, thus mediating the CXCL12-dependent recruitmentf regenerative CXCR4+ progenitor cells [25,26].

In addition to CXCR4, EPCs also express the CXC chemokineeceptor-2 (CXCR2), and ligands (CXCL1 and CXCL7) for those recep-

ors were consecutively up regulated during inflammation but alsofter arterial injury, which may ameliorate endothelial recoveryy selectively recruiting myeloid CD14+/low EPC subsets [14,27].ccordingly, blockade of the angiogenic CXCR2 ligand keratinocyte-erived chemokine (KC/CXCL1) inhibited endothelial recovery and

dmac

l Research 58 (2008) 148–151 149

ncreased neointimal growth in Apoe−/− mice after wire-injury ofhe carotid artery [27]. Moreover, blocking the pro-inflammatory

acrophage migration inhibitory factor (MIF) associated with aonversion into a more stable plaque phenotype and severelyeduced macrophage infiltration [28]. This could be explained byhe finding that MIF acts as a dual agonist of both, CXCR2 andXCR4, resulting in recruitment of macrophages but possibly alsoascular progenitor cells, which may balance their contributiono neointimal area [29]. In contrast, MIF is expected to retain aotent and pre-dominant CXCR2 activity in primary atherogene-is, amounting to regression of established plaques. Parallel to CXChemokine/receptor pairs, the CC chemokine/receptor couples alsolay a central role in the homing of EPCs after arterial injury [30,31].or instance, infused bone marrow myeloid cells over-expressingCL2 significantly accelerated endothelial healing and reduced theeointima thus unveiling the importance of the CCL2/CCR2 axis31]. Constitutively active beta-integrins and the P-selectin glyco-rotein ligand-1 were also crucially involved in the recruitment ofPCs in vitro as well as in vivo [32–34]. Last but not least, activatedlatelets at the site of arterial injury have been shown to supportrrest and differentiation of circulating EPCs [30].

Nitric oxide was identified as another powerful and specificobilizing factor for EPCs and their recruitment was consequently

mpaired in eNos−/− mice [35]. In addition, hypoxia and someytokines (e.g. VEGF, erythropoietin and G-CSF) have been showno increase the number of EPCs [15,36,37]. Similar to hypoxia inhe context of vascular damage, burn injury or surgical bypassntervention rapidly mobilized circulating CD133+VEGFR2+ EPCs38]. CD34+ cell subsets were differentially affected in patientsith heart failure, acute myocardial infarction or chronic coro-ary endothelial dysfunction [39–42]. Even more physiologically,strogens and physical training were reported to associate withlevated EPC counts [9]. Therapeutically, statins and thiazolidin-iones transiently mobilized circulating EPCs but also facilitatedheir neo-endothelial incorporation, thus suppressing neointi-

al hyperplasia [43–45]. Conversely, long-term treatment withtatins dose dependently decreased the number of circulatingD34+VEGFR2+ EPCs but increased another subpopulation oftill more mature CD34+CD144+ cells in the systemic circulation46–48]. Recent evidence further revealed positive effects of somenti-hypertensive therapeutics such as calcium antagonists andngiotensin-converting enzyme inhibitors on number and functionf EPCs [49,50].

. Involvement of EPCs in vascular repair and remodeling

Endothelial dysfunction is an early hallmark of atheroscleroticisease and may correlate to ischemic events even in the absence ofrterial obstruction. In the context of regeneration, published datarom animal studies have unveiled that EPCs effectively contributen restoring endothelial function and diminishing neointimal for-

ation after arterial injury [14,20,30,44]. EPCs have also beenhown to participate in regeneration of damaged endothelial cellsn atherosclerosis-prone Apoe−/− mice and a large percentage ofenewed endothelial cells in vascular grafts originated from circu-ating progenitors [51,52]. Parallel to these findings EPCs were alsorucially involved in neovascularization of ischemic tissue by cre-ting new vessels and by delivering of angiogenic growth factors8,9].

Translating this basic knowledge to the clinical setting has intro-uced the therapeutic application of autologous EPCs or bonearrow mononuclear cells for adjuvant cell-based treatment of

cute as well chronic myocardial ischemia [53]. The commonlinical concept currently claims an even protective role of EPCs

Page 3: Endothelial progenitor cells in vascular repair and remodeling

150 M. Hristov, C. Weber / Pharmacologica

Table 1Involvement of EPCs in primary atherosclerosis and injury repair

Stage of arterial disease Cellular effect and mechanism

Primary atherosclerosisEarly lesions (endothelial dysfunction) Protective endothelial recoveryPlaque progression Incorporation at predeliction

sitesAdvanced rupture-prone plaques Neovascularization,

destabilization

Secondary atherosclerosis after injuryRepair after plaque rupture Possible contribution to

regenerationBalloon injury/stent and transplant Reendothelialization and

dtrFatbwebeFialaTcitid

5

titawliohritf

R

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

reduced neointimaMyocardial ischemia and infarction Mobilization and contribution

to tissue repair

uring the progression of atherosclerosis and further suggests thathese cells may serve as reliable biomarker for endogenous vascularepair [54]. Reduced numbers of EPCs have been associated with theramingham risk factor score, peripheral endothelial dysfunctionnd the incidence of future cardiovascular events [55,56]. Of note,he number of EPCs significantly increased in patients with unsta-le angina, while their function did not differ as compared to thoseith stable coronary artery disease [57]. Some recent studies, how-

ver, revealed that the number of circulating EPCs was not affectedy cardiovascular risk factors but obviously associated with thextent of vessel disease and long-term statin therapy [41,46,58,59].urthermore, published data in animal models have shown thatnfusion of EPCs increased lipid content and decreased collagenmount in atherosclerotic plaques of Apoe−/− mice [60]. In the sameine elevated levels of plasma CXCR2 receptor ligands such as CXCL1nd CXCL7 were clinically related to plaque destabilization [61,62].hese findings may be explained in part by influx of CXCR2+ mono-yte subsets that include also putative endothelial precursors withnflammatory, proteolytic and angiogenic properties [14,63]. Thus,he dual contribution of EPC subsets to vascular remodeling dur-ng generalized advanced atherosclerosis versus local endothelialysfunction after arterial injury need a critical re-evaluation.

. Conclusions

Further to chronic inflammation and immunological interac-ions, vascular progenitor cells have been recognized as pivotallymplicated players in the pathogenesis of atherosclerosis. In par-icular, EPCs obviously participate in endothelial maintenancend their number is affected during coronary artery disease. Aidespread EPC mobilization may associate with plaque destabi-

ization, whereas local stimulation of EPC recruitment after arterialnjury may even effectively support endothelial healing. Basedn this ambivalence, selectively controlling the mobilization andoming of EPCs will help to regulate the endogenous arterialegenerative potential (Table 1). This knowledge appears crucialn the clinical application of EPCs in terms of reliable prognos-ic/diagnostic biomarker but also of devising therapeutic targetsor cardiovascular regenerative medicine.

eferences

[1] Cines DB, Pollak ES, Buck CA, Loscalzo J, Zimmerman GA, McEver RP, et al.

Endothelial cells in physiology and in the pathophysiology of vascular disor-ders. Blood 1998;91:3527–61.

[2] Dignat-George F, Sampol J. Circulating endothelial cells in vascular disorders:new insights into an old concept. Eur J Haematol 2000;65:215–20.

[3] Hansson GK, Inflammation. atherosclerosis, and coronary artery disease. N EnglJ Med 2005;352:1685–95.

[

[

l Research 58 (2008) 148–151

[4] Libby P, Aikawa M. Stabilization of atherosclerotic plaques: new mechanismsand clinical targets. Nat Med 2002;8:1257–62.

[5] Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitorendothelial cells for angiogenesis. Science 1997;275:964–7.

[6] Peichev M, Naiyer AJ, Pereira D, et al. Expression of VEGFR-2 and AC133 by cir-culating human CD34(+) cells identifies a population of functional endothelialprecursors. Blood 2000;95:952–8.

[7] Yoon YS, Wecker A, Heyd L, et al. Clonally expanded novel multipotent stem cellsfrom human bone marrow regenerate myocardium after myocardial infarction.J Clin Invest 2005;115:326–38.

[8] Rabelink TJ, de Boer HC, de Koning EJ, van Zonneveld AJ. Endothelial progeni-tor cells: more than an inflammatory response? Arterioscler Thromb Vasc Biol2004;24:834–8.

[9] Hristov M, Weber C. Ambivalence of progenitor cells in vascular repair andplaque stability. Curr Opin Lipidol; in press.

10] Zengin E, Chalajour F, Gehling UM, et al. Vascular wall resident progenitor cells:a source for postnatal vasculogenesis. Development 2006;133:1543–51.

11] Harraz M, Jiao C, Hanlon HD, Hartley RS, Schatteman GC. CD34-blood-derivedhuman endothelial cell progenitors. Stem Cells 2001;19:304–12.

12] Elsheikh E, Uzunel M, He Z, et al. Only a specific subset of humanperipheral-blood monocytes has endothelial-like functional capacity. Blood2005;106:2347–55.

13] Venneri MA, De Palma M, Ponzoni M, et al. Identification of proangiogenic TIE2-expressing monocytes (TEMs) in human peripheral blood and cancer. Blood2007;109:5276–85.

14] Hristov M, Zernecke A, Bidzhekov K, et al. Importance of CXC chemokine recep-tor 2 in the homing of human peripheral blood endothelial progenitor cells tosites of arterial injury. Circ Res 2007;100:590–7.

15] Kalka C, Masuda H, Takahashi T, Gordon R, Tepper O, Gravereaux E, etal. Vascular endothelial growth factor165 gene transfer augments circulat-ing endothelial progenitor cells in human subjects. Circ Res 2000;86:1198–202.

16] Rehman J, Li J, Orschell CM, March KL. Peripheral blood “endothelial progenitorcells” are derived from monocyte/macrophages and secrete angiogenic growthfactors. Circulation 2003;107:1164–9.

17] Lin Y, Weisdorf DJ, Solovey A, Hebbel RP. Origins of circulating endothelial cellsand endothelial outgrowth from blood. J Clin Invest 2000;105:71–7.

18] Gulati R, Jevremovic D, Peterson TE, Chatterjee S, Shah V, Vile RG, et al. Diverseorigin and function of cells with endothelial phenotype obtained from adulthuman blood. Circ Res 2003;93:1023–5.

19] Yoder MC, Mead LE, Prater D, Krier TR, Mroueh KN, Li F, et al. Redefin-ing endothelial progenitor cells via clonal analysis and hematopoieticstem/progenitor cell principals. Blood 2007;109:1801–9.

20] Wang CH, Cherng WJ, Yang NI, et al. Late-outgrowth endothelial cells attenu-ate intimal hyperplasia contributed by mesenchymal stem cells after vascularinjury. Arterioscler Thromb Vasc Biol 2008;28:54–60.

21] Petit I, Jin D, Rafii S. The SDF-1-CXCR4 signaling pathway: a molecular hubmodulating neo-angiogenesis. Trends Immunol 2007;28:299–307.

22] Heissig B, Hattori K, Dias S, et al. Recruitment of stem and progenitor cells fromthe bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell2002;109:625–37.

23] Schober A, Karshovska E, Zernecke A, Weber C. SDF-1�-mediated tissue repairby stem cells: a promising tool in cardiovascular medicine? Trends CardiovascMed 2006;16:103–8.

24] Damås JK, Waehre T, Yndestad A, et al. Stromal cell-derived factor-1alpha inunstable angina: potential antiinflammatory and matrix-stabilizing effects. Cir-culation 2002;106:36–42.

25] Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. Progenitor cell trafficking isregulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med2004;10:858–64.

26] Karshovska E, Zernecke A, Sevilmis G, et al. Expression of HIF-1� in injuredarteries controls SDF-1alpha mediated neointima formation in apolipoproteinE deficient mice. Arterioscler Thromb Vasc Biol 2007;27:2540–7.

27] Liehn EA, Schober A, Weber C. Blockade of keratinocyte-derived chemokineinhibits endothelial recovery and enhances plaque formation after arte-rial injury in ApoE-deficient mice. Arterioscler Thromb Vasc Biol 2004;24:1891–6.

28] Schober A, Bernhagen J, Thiele M, et al. Stabilization of atherosclerotic plaquesby blockade of macrophage migration inhibitory factor after vascular injury inapolipoprotein E-deficient mice. Circulation 2004;109:380–5.

29] Bernhagen J, Krohn R, Lue H, et al. MIF is a noncognate ligand of CXCchemokine receptors in inflammatory and atherogenic cell recruitment. NatMed 2007;13:587–96.

30] Hristov M, Zernecke A, Liehn EA, Weber C. Regulation of endothelial progenitorcell homing after arterial injury. Thromb Haemost 2007;98:274–7.

31] Fujiyama S, Amano K, Uehira K, et al. Bone marrow monocyte lineage cellsadhere on injured endothelium in a monocyte chemoattractant protein-1-dependent manner and accelerate reendothelialization as endothelialprogenitor cells. Circ Res 2003;93:980–9.

32] Chavakis E, Aicher A, Heeschen C, et al. Role of �2-integrins for homingand neovascularization capacity of endothelial progenitor cells. J Exp Med2005;201:63–72.

33] Duan H, Cheng L, Sun X, et al. LFA-1 and VLA-4 involved in human high prolifer-ative potential-endothelial progenitor cells homing to ischemic tissue. ThrombHaemost 2006;96:807–15.

Page 4: Endothelial progenitor cells in vascular repair and remodeling

ologica

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

M. Hristov, C. Weber / Pharmac

34] Foubert P, Silvestre JS, Souttou B, et al. PSGL-1-mediated activation of EphB4increases the proangiogenic potential of endothelial progenitor cells. J ClinInvest 2007;117:1527–37.

35] Aicher A, Heeschen C, Mildner-Rihm C, et al. Essential role of endothelialnitric oxide synthase for mobilization of stem and progenitor cells. Nat Med2003;9:1370–6.

36] Bahlmann FH, De Groot K, Spandau JM, et al. Erythropoietin regulates endothe-lial progenitor cells. Blood 2004;103:921–6.

37] Honold J, Lehmann R, Heeschen C, et al. Effects of granulocyte colonysimulating factor on functional activities of endothelial progenitor cells inpatients with chronic ischemic heart disease. Arterioscler Thromb Vasc Biol2006;26:2238–43.

38] Gill M, Dias S, Hattori K, et al. Vascular trauma induces rapid but tran-sient mobilization of VEGFR2(+)AC133(+) endothelial precursor cells. Circ Res2001;88:167–74.

39] Valgimigli M, Rigolin GM, Fucili A, Porta MD, Soukhomovskaia O, Malagutti P,et al. CD34+ and endothelial progenitor cells in patients with various degreesof congestive heart failure. Circulation 2004;110:1209–12.

40] Shintani S, Murohara T, Ikeda H, Ueno T, Honma T, Katoh A, et al. Mobilizationof endothelial progenitor cells in patients with acute myocardial infarction.Circulation 2001;103:2776–9.

41] Massa M, Rosti V, Ferrario M, Campanelli R, Ramajoli I, Rosso R, et al. Increasedcirculating hematopoietic and endothelial progenitor cells in the early phaseof acute myocardial infarction. Blood 2005;105:199–206.

42] Boilson BA, Kiernan TJ, Harbuzariu A, Nelson RE, Lerman A, Simari RD. Circulat-ing CD34+ cell subsets in patients with coronary endothelial dysfunction. NatClin Pract Cardiovasc Med 2008;5:489–96.

43] Vasa M, Fichtlscherer S, Adler K, et al. Increase in circulating endothelial pro-genitor cells by statin therapy in patients with stable coronary artery disease.Circulation 2001;103:2885–90.

44] Walter DH, Rittig K, Bahlmann FH, et al. Statin therapy accelerates reendothe-lialization: a novel effect involving mobilization and incorporation of bonemarrow-derived endothelial progenitor cells. Circulation 2002;105:3017–24.

45] Gensch C, Clever YP, Werner C, Hanhoun M, Böhm M, Laufs U. The PPAR-gamma agonist pioglitazone increases neoangiogenesis and prevents apoptosisof endothelial progenitor cells. Atherosclerosis 2007;192:67–74.

46] Hristov M, Fach C, Becker C, et al. Reduced numbers of circulating endothelialprogenitor cells in patients with coronary artery disease associated with long-term statin treatment. Atherosclerosis 2007;192:413–20.

47] Deschaseaux F, Selmani Z, Falcoz PE, et al. Two types of circulating endothelial

progenitor cells in patients receiving long term therapy by HMG-CoA reductaseinhibitors. Eur J Pharmacol 2007;562:111–8.

48] Redondo S, Hristov M, Gordillo-Moscoso AA, et al. High-reproducible flowcytometric endothelial progenitor cell determination in human peripheralblood as CD34+/CD144+/CD3− lymphocyte sub-population. J Immunol Method2008;335:21–7.

[

l Research 58 (2008) 148–151 151

49] Sugiura T, Kondo T, Kureishi-Bando Y, Numaguchi Y, Yoshida O, Dohi Y, KimuraG, Ueda R, Rabelink TJ, Murohara T. Nifedipine improves endothelial function.Role of endothelial progenitor cells. Hypertension 2008;52:1–8.

50] Wang CH, Verma S, Hsieh IC, Chen YJ, Kuo LT, Yang NI, et al. Enalapril increasesischemia-induced endothelial progenitor cell mobilization through manipula-tion of the CD26 system. J Mol Cell Cardiol 2006;41:34–43.

51] Rauscher FM, Goldschmidt-Clermont PJ, Davis BH, Wang T, Gregg D,Ramaswami P, et al. Aging, progenitor cell exhaustion, and atherosclerosis.Circulation 2003;108:457–63.

52] Hu Y, Davison F, Zhang Z, Xu Q. Endothelial replacement and angiogenesis inarteriosclerotic lesions of allografts are contributed by circulating progenitorcells. Circulation 2003;108:3122–7.

53] Hristov M, Weber C. The therapeutic potential of progenitor cells in ischemicheart disease—past, present and future. Basic Res Cardiol 2006;101:1–7.

54] Leor J, Marber M. Endothelial progenitors: a new Tower of Babel? J Am CollCardiol 2006;48:1588–90.

55] Werner N, Nickenig G. Influence of cardiovascular risk factors on endothe-lial progenitor cells: limitations for therapy? Arterioscler Thromb Vasc Biol2006;26:257–66.

56] Murphy C, Kanaganayagam GS, Jiang B, Chowienczyk PJ, Zbinden R, Saha M,et al. Vascular dysfunction and reduced circulating endothelial progenitorcells in young healthy UK South Asian men. Arterioscler Thromb Vasc Biol2007;27:936–42.

57] George J, Goldstein E, Abashidze S, Deutsch V, Shmilovich H, Finkelstein A, etal. Circulating endothelial progenitor cells in patients with unstable angina:association with systemic inflammation. Eur Heart J 2004;25:1003–8.

58] Güven H, Shepherd RM, Bach RG, et al. The number of endothelial progenitor cellcolonies in the blood is increased in patients with angiographically significantcoronary artery disease. J Am Coll Cardiol 2006;48:1579–87.

59] Xiao Q, Kiechl S, Patel S, et al. Endothelial progenitor cells, cardiovascular riskfactors, cytokine levels and atherosclerosis—results from a large population-based study. PLoS ONE 2007;2:e975.

60] George J, Afek A, Abashidze A, et al. Transfer of endothelial progenitorand bone marrow cells influences atherosclerotic plaque size and compo-sition in apolipoprotein E knockout mice. Arterioscler Thromb Vasc Biol2005;25:2636–41.

61] Smith C, Damås JK, Otterdal K, et al. Increased levels of neutrophil-activatingpeptide-2 in acute coronary syndromes: possible role of platelet-mediatedvascular inflammation. J Am Coll Cardiol 2006;48:1591–9.

62] Breland UM, Halvorsen B, Hol J, et al. A potential role of the CXC chemokine

GRO� in atherosclerosis and plaque destabilization. Downregulatory effects ofstatins. Arterioscler Thromb Vasc Biol 2008;28:1005–11.

63] Boisvert WA, Santiago R, Curtiss LK, Terkeltaub RA. A leukocyte homologueof the IL-8 receptor CXCR-2 mediates the accumulation of macrophagesin atherosclerotic lesions of LDL receptor-deficient mice. J Clin Invest1998;101:353–63.