+ All Categories
Home > Documents > Cytokine-Induced Mobilization of Circulating Endothelial Progenitor Cells Enhances Repair of Injured...

Cytokine-Induced Mobilization of Circulating Endothelial Progenitor Cells Enhances Repair of Injured...

Date post: 06-Mar-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
9
Chong C. Liew, Richard E. Pratt and Victor J. Dzau Deling Kong, Luis G. Melo, Massimiliano Gnecchi, Lunan Zhang, Gustavo Mostoslavsky, Repair of Injured Arteries Cytokine-Induced Mobilization of Circulating Endothelial Progenitor Cells Enhances Print ISSN: 0009-7322. Online ISSN: 1524-4539 Copyright © 2004 American Heart Association, Inc. All rights reserved. is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231 Circulation doi: 10.1161/01.CIR.0000143161.01901.BD 2004;110:2039-2046; originally published online September 27, 2004; Circulation. http://circ.ahajournals.org/content/110/14/2039 World Wide Web at: The online version of this article, along with updated information and services, is located on the http://circ.ahajournals.org//subscriptions/ is online at: Circulation Information about subscribing to Subscriptions: http://www.lww.com/reprints Information about reprints can be found online at: Reprints: document. Permissions and Rights Question and Answer this process is available in the click Request Permissions in the middle column of the Web page under Services. Further information about Office. Once the online version of the published article for which permission is being requested is located, can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial Circulation in Requests for permissions to reproduce figures, tables, or portions of articles originally published Permissions: by guest on November 13, 2013 http://circ.ahajournals.org/ Downloaded from by guest on November 13, 2013 http://circ.ahajournals.org/ Downloaded from
Transcript

Chong C. Liew, Richard E. Pratt and Victor J. DzauDeling Kong, Luis G. Melo, Massimiliano Gnecchi, Lunan Zhang, Gustavo Mostoslavsky,

Repair of Injured ArteriesCytokine-Induced Mobilization of Circulating Endothelial Progenitor Cells Enhances

Print ISSN: 0009-7322. Online ISSN: 1524-4539 Copyright © 2004 American Heart Association, Inc. All rights reserved.

is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231Circulation doi: 10.1161/01.CIR.0000143161.01901.BD

2004;110:2039-2046; originally published online September 27, 2004;Circulation. 

http://circ.ahajournals.org/content/110/14/2039World Wide Web at:

The online version of this article, along with updated information and services, is located on the

  http://circ.ahajournals.org//subscriptions/

is online at: Circulation Information about subscribing to Subscriptions: 

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

  document. Permissions and Rights Question and Answer this process is available in the

click Request Permissions in the middle column of the Web page under Services. Further information aboutOffice. Once the online version of the published article for which permission is being requested is located,

can be obtained via RightsLink, a service of the Copyright Clearance Center, not the EditorialCirculationin Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions:

by guest on November 13, 2013http://circ.ahajournals.org/Downloaded from by guest on November 13, 2013http://circ.ahajournals.org/Downloaded from

Cytokine-Induced Mobilization of Circulating EndothelialProgenitor Cells Enhances Repair of Injured Arteries

Deling Kong, PhD; Luis G. Melo, PhD; Massimiliano Gnecchi, MD; Lunan Zhang, MD;Gustavo Mostoslavsky, MD, PhD; Chong C. Liew, PhD; Richard E. Pratt, PhD; Victor J. Dzau, MD

Background—The existence of circulating endothelial progenitor cells (CEPCs) has previously been documented. Thesecells can be mobilized by cytokines and are recruited to sites of injury, where they may participate in tissue repair. Inthe present study, we examined the hypothesis that mobilization of CEPCs by exogenous granulocyte-colony stimulatingfactor (G-CSF) enhances repair of injured arteries by facilitating reendothelialization and inhibiting neointimadevelopment.

Methods and Results—Male rats were injected daily with 50 �g/kg recombinant human G-CSF or 0.9% NaCl SC for 8days. On the fifth day of treatment, 1 mL of blood was collected for fluorescence-activated cell sorting analysis ofmononuclear cells, and the animals underwent balloon angioplasty of the common carotid artery. The animals werekilled at 2 or 4 weeks after injury, and the carotid arteries were harvested and processed for immunohistochemistry,scanning electron microscopy (SEM), and morphometric analysis of endothelialization and neointimal formation.G-CSF increased the number of circulating mononuclear cells that express endothelial cell lineage markers several-fold.SEM and immunohistochemical staining with the endothelial marker, platelet and endothelial cell adhesion molecule-1,showed rapid and nearly complete (�90%) reendothelialization of the denuded vessels in the G-CSF–treated animalscompared with �20% in the control animals. Reendothelialization was paralleled by a decrease in inflammation in thevessel wall. Neointima thickness was reduced by �60% in the G-CSF–treated animals compared with control animalsat 2 and 4 weeks after injury.

Conclusion—We postulate that cytokine-induced mobilization of CEPCs may be a suitable therapeutic strategy forprevention of restenosis after revascularization procedures. (Circulation. 2004;110:2039-2046.)

Key Words: growth substances � restenosis � endothelium � immunohistochemistry

Vascular injury leads to pathological repair and remodel-ing that involve vascular smooth muscle cell migration

and proliferation, resulting in neointimal hyperplasia.1 Endo-thelial cell (EC) loss is a major contributing factor to thepathological repair of the injured blood vessel.2 The disrup-tion of endothelial integrity leads to a concomitant reductionin the production of vasculoprotective mediators, such asnitric oxide and prostacyclin, and increased vasoconstrictorand growth-promoting substances,3,4 resulting in elevatedvascular tone, platelet adhesion, enhanced inflammation, andmedial smooth muscle cell proliferation.3,4 The resultantneointimal hyperplasia is the pathological basis for restenosisafter revascularization procedures such as angioplasty, stent-ing, and bypass grafting.1,2

Because EC loss plays a pivotal role in the pathogenesis ofintimal hyperplasia after vascular injury, we postulated that atherapeutic strategy that promotes early reendothelializationof the injured vessels would inhibit intimal lesion develop-

ment, facilitate vascular repair, and improve long-term vesselpatency. Progenitor cells originating from the bone marrowhave previously been isolated from the mononuclear cellfraction of peripheral blood.5,6 These cells have high prolif-erative potential5 and under specific growth conditions, dif-ferentiate into ECs,7 suggesting that they may be suitable asa substrate for the reendothelialization of damaged vessels.We showed recently that transplantation of autologous circu-lating endothelial progenitor cells (CEPCs) onto balloon-denuded arteries led to rapid reendothelialization of theinjured artery.8 Others have reported that transplantation ofCEPCs onto the decellularized porcine iliac artery led toformation of a bioactive endothelial monolayer and pro-longed graft patency when these preparations were implantedas carotid interposition grafts.9

We hypothesized that mobilization of CEPCs by hemato-poietic growth factors such as granulocyte-colony stimulatingfactor (G-CSF) may provide a potentially effective noninva-

Received October 21, 2003; de novo received February 28, 2004; revision received May 17, 2004; accepted May 20, 2004.From the Department of Medicine (D.K., L.G.M., M.G., L.Z., C.C.L., R.E.P., V.J.D.), Brigham and Women’s Hospital and Harvard Medical School,

Boston, Mass; the Department of Physiology (L.G.M.), Queen’s University, Kingston, Ontario, Canada; and the Department of Genetics (G.M.),Children’s Hospital, Boston, Mass. Dr Kong is currently at the School of Life Science, Nankai University, Tianjin, China.

Correspondence to Victor J. Dzau, MD, Office of the Chancellor, Duke University Medical Center, DUMC 3701, Durham, NC 27710. [email protected]

© 2004 American Heart Association, Inc.

Circulation is available at http://www.circulationaha.org DOI: 10.1161/01.CIR.0000143161.01901.BD

2039 by guest on November 13, 2013http://circ.ahajournals.org/Downloaded from

sive strategy to enhance reendothelialization of injured ves-sels. Several studies have shown that exogenous administra-tion of cytokines increases the number of CEPCs.10–12 Forexample, pretreatment with vascular endothelial growth fac-tor was reported to double the number of circulating CEPCsin humans,10,11 and administration of G-CSF and stem cellfactor was found to mobilize EPCs from the bone marrow.11

Recently, Bhattacharya et al12 and Shi et al13 reported thatG-CSF enhances endothelialization of small-caliber prosthet-ic implanted grafts, whereas Walter el al14 showed a compa-rable effect in injured carotid arteries of mice after statintherapy for several weeks before injury.

In the present study, we evaluated the efficacy of short-term G-CSF pretreatment as a strategy for promoting reen-dothelialization and inhibition of neointimal hyperplasia ininjured arteries. We chose to use G-CSF because of its abilityto efficiently mobilize hematopoietic precursor cells from thebone marrow15 and to stimulate EC migration and prolifera-tion.16 Our results show that short-term treatment with G-CSFbefore balloon angioplasty leads to accelerated reendotheli-alization and marked inhibition of neointimal formation in theinjured vessels.

MethodsAnimalsMale Sprague-Dawley rats (200 to 250 g body weight) werepurchased from Harlan Laboratories (Indianapolis, Ind). The animalswere maintained on a 12-hour:12-hour light/dark cycle under ambi-ent conditions of 24°C and 60% humidity. Food and water wereprovided ad libitum. The Harvard Medical Area Standing Committeeon Animal Care approved all animal procedures.

Cytokine-Induced Mononuclear Cell MobilizationThe animals were anesthetized by intraperitoneal injection withketamine (70 mg/kg) and xylazine (4 mm/kg) and splenectomized toeliminate the spleen as a source of CEPCs. Two weeks aftersplenectomy, the animals were injected daily with 50 �g/kg SCrecombinant human G-CSF (a generous gift from Dr Pamela Baker,University of Massachusetts, Amherst) for 8 days. Control animalswere treated with an equivalent volume of saline.

Balloon-Injury ModelBalloon injury of the left common carotid artery was performed onthe fifth day after initiation of G-CSF treatment, as previouslydescribed.8 In brief, a 2F Fogarty arterial embolectomy catheter(Edwards Lifesciences) was inserted through the external carotidartery, inflated with 200 �L air, and passed 3 times along the lengthof the segment.

Figure 1. FACS analysis of cytokine-induced mononuclear cell mobilization in splenectomized rats. A, Mononuclear cell mobilization byG-CSF. B, Percentage of mononuclear cells coexpressing hematopoietic and endothelial lineage markers CD45 and CD31. C, Percent-age of mononuclear cells coexpressing endothelial lineage markers CD34, endothelial nitric oxide synthase (eNOS), Flk-1, stem cell fac-tor receptor c-kit, and E-selectin. D, FACS histograms of hematopoietic and EC markers. *P�0.05 saline vs G-CSF. PE indicates phy-coerythrin; FITC, fluorescein isothiocyanate; eGFP, enhanced green fluorescence protein; and APC, allophycocyanin. All otherabbreviations are as defined in text.

2040 Circulation October 5, 2004

by guest on November 13, 2013http://circ.ahajournals.org/Downloaded from

Characterization of Cytokine-MobilizedMononuclear CellsOn the fifth day of G-CSF or saline treatment, 1 mL blood washarvested from the inferior vena cava of each rat. The mononuclear cellswere isolated by density-gradient centrifugation8 and counted with a Z1Coulter particle counter (Beckman Coulter). Approximately 106 cellsfrom each animal were suspended in 50 �L phosphate-buffered salinecontaining 5 mmol/L EDTA and 0.5% bovine serum albumin andincubated for 30 minutes on ice with 20 �g/mL phycoerythrin-conjugated anti-mouse Flk-1, allophycocyanin-conjugated anti-mousec-kit, and fluorescein isothiocyanate–conjugated anti-mouse CD45 oranti-mouse vascular endothelial (VE)-cadherin. All primary antibodieswere purchased from BD Pharmingen with the exception of VE-cadherin, which was purchased from R&D Systems. Unlabeled cellsserved as negative controls. The cells were analyzed by 3-colorfluorescence activated cell sorting (FACS) with a FACSCalibur flowcytometer (Beckman Dickinson) equipped with a 488-nm and a 635-nmred diode laser. Propidium iodide staining was used to gate out deadcells.

ImmunohistochemistryCarotid arteries were harvested 7 and 14 days after injury and frozenin OCT compound (Miles). Sections (5 �m) were incubated in a 1:50dilution of anti-rat monoclonal CD-31 (platelet and endothelial celladhesion molecule-1, BD Biosciences). Adjacent sections wereincubated in 1:50 monoclonal anti-rat vascular cell adhesionmolecule-1 (VCAM-1), 1:100 polyclonal anti-rat CD45 (BD Bio-sciences), or monoclonal rat endothelial cell antibody (RECA-1,Abcam). All sections were then incubated in a 1:400 dilution ofbiotinylated anti-mouse rat IgG (Vector Laboratories), treated withalkaline phosphatase, and developed in Sigma Fast Red (Sigma). Thesections were counterstained with hematoxylin.

Morphometric Analysis of Endothelialization andNeointimal HyperplasiaMorphometric assessment of endothelialization and neointimal hy-perplasia was performed in frozen sections 2 and 4 weeks afterinjury, as previously described.8 The integrity of the reconstitutedendothelium was also verified by scanning electron microscopy(SEM). For morphometric analysis of neointimal hyperplasia, 6 to 8individual, elastin-stained, paraffin sections sampled from 4 differentregions of the injured segment were used to calculate neointimal andmedial thicknesses, cross-sectional areas, and luminal diameter withthe use of ImagePro software.

Statistical AnalysisAll results are presented as mean�SEM. An unpaired t test was usedfor comparisons between control and treated groups. One-way

ANOVA followed by Bonferroni multiple-comparison test was usedfor comparisons between groups at different time points. A proba-bility value �0.05 was considered to indicate statistical significance.

ResultsHematopoietic Cell Mobilization and FACS AnalysisThe effect of G-CSF treatment on hematopoietic cell mobi-lization is shown in Figure 1A. After 5 days of G-CSFtreatment, the number of circulating mononuclear cells in-creased by 2-fold (control, 5.96�0.35�106 cells; G-CSF,13.07�1.376�106 cells; P�0.001, n�5 for each group).FACS analysis of the whole mononuclear fraction at this timeshowed a 3.3-fold increase in the percentage of mononuclearcells expressing the common leukocyte antigen CD45 (con-trol, 2.84�106; G-CSF, 9.41�106 cells/mL blood; Figure 1Band 1D). Comparable numbers were seen for cells expressingCD31, a surface molecule in ECs (Figure 1B and 1D).Approximately 6% of the cells expressed the hematopoieticlineage marker CD34 in the G-CSF–treated group comparedwith 2% in the saline control, corresponding to a 7-foldincrease in the number of positive cells (control, 1.2�105;G-CSF, 8.4�105 cells; Figure 1C and 1D). The endothelialmarkers endothelial nitric oxide synthase, Flk1, andE-selectin were expressed in 8.7%, 6.1%, and 2.7% of themononuclear cells from G-CSF–treated animals comparedwith 1.3%, 3.0%, and 2.5%, respectively, of the controls,corresponding to an �15-, 4.5-, and 2.3-fold increase in thenumber of cells expressing these markers (Figure 1C and 1D).Approximately 2.4�105 cells expressed the stem cell markerc-kit in the G-CSF–mobilized group compared with 8.7�104

in the control group (Figure 1C and 1D). These resultssuggest that G-CSF treatment significantly increases thenumber of circulating cells expressing an endothelial lineagephenotype.

Balloon Injury and InflammationThe morphology of the intact and balloon-injured carotidvessel wall is shown in Figure 2. Histological analysis ofhematoxylin-and-eosin–stained cross sections and topo-graphic SEM of the luminal surface of uninjured vessels

Figure 2. Morphology of commoncarotid artery after balloon injury. A,Hematoxylin-and-eosin–stained sectionfrom uninjured vessel. Arrows indicatepresence of endothelial monolayer. B,Topographic SEM photomicrograph ofuninjured vessel, showing monolayer ofdensely packed ECs (arrows). C, Hema-toxylin-and-eosin–stained section fromballoon-injured carotid artery. Arrowsshow absence of endothelium. D, SEMphotomicrograph of injured vessel,showing exposed subendothelial fibersand absence of ECs. Histological sec-tions were viewed at �400. SEM sam-ples were viewed at �1000. All abbrevia-tions are as defined in text.

Kong et al Bone Marrow Mobilization and Neointima Formation 2041

by guest on November 13, 2013http://circ.ahajournals.org/Downloaded from

showed a continuous endothelial monolayer lining the vessellumen (Figure 2A and 2B). The integrity of the endotheliumwas further confirmed by immunohistochemical detection ofRECA-1 (data not shown). The angioplasty procedure led tonearly complete disruption of the endothelium, as evidencedby the presence of an interrupted intima exposing the suben-dothelial fibers (Figure 2C and 2D). Intense VCAM-1 immu-noreactivity was detected in the medial and intimal layers ofcarotid vessels from control animals on day 4 after injury(Figure 3A) but was markedly reduced in G-CSF–treatedanimals (Figure 3B). After 2 weeks, VCAM-1 activity wasstill detectable in the control sections, albeit at a lowerintensity (Figure 3C). In contrast, no VCAM-positive immu-noreactivity was present in the sections from G-CSF–treatedanimals (Figure 3D). A small number of CD45-positive cellswas found in the adventitia in saline-treated (Figure 3E and3G) and G-CSF–treated (Figure 3F and 3H) animals, but noleukocyte infiltration was seen in the intima at either timepoint.

Reendothelialization of Denuded Carotid ArteryWe assessed the effect of G-CSF treatment on reendotheli-alization of denuded vessels in sections stained with CD31

and RECA-1 14 days after injury, as well as by SEM.Sections from control animals showed patchy and interruptedCD31- (Figure 4A) and RECA-1– (Figure 4B) positivestaining. A topographic SEM view of the untreated vesselsshowed incomplete and sparse endothelium (Figure 4C). Incontrast, a nearly complete and continuous monolayer ofCD31- (Figure 4D) and RECA-1– (Figure 4E) positive cellswas found lining the lumen of G-CSF–treated animals. SEMof the luminal surface of these vessels revealed the presenceof densely packed, continuous endothelium-like cells cover-ing the luminal area (Figure 3F). Morphometric analysis ofthe CD31-positive area showed �90% EC coverage of thelumen in the G-CSF–treated animals compared with �20% inthe control animals (Figure 4G).

Inhibition of Neointimal HyperplasiaWe determined the ability of G-CSF pretreatment to inhibitneointimal hyperplasia of the balloon-injured carotid vesselsat 2 and 4 weeks after injury. Two weeks after injury, aprominent, concentric neointima had developed in the un-treated vessels (Figure 5A). By 4 weeks the neointima hadslightly expanded, but this did not reach statistical signifi-

Figure 3. Inflammation in vessel wallafter balloon injury. A and B, Immunohis-tochemical staining of VCAM-1 4 daysafter balloon injury in untreated andG-CSF–treated animals, respectively. Cand D, VCAM-1 staining 14 days afterinjury in untreated and G-CSF–treatedanimals, respectively. Arrows indicateVCAM-1–positive staining. E and F,CD45 immunostaining 4 days after injuryin saline- and G-CSF–treated animals,respectively. G and H, CD45 immuno-staining 14 days after injury. Arrows indi-cate CD45-positive cells. Sections werecounterstained with hematoxylin andviewed at �400. All abbreviations are asdefined in text.

2042 Circulation October 5, 2004

by guest on November 13, 2013http://circ.ahajournals.org/Downloaded from

cance relative to 2 weeks (Figure 5B). Neointimal formationwas markedly reduced in the G-CSF–treated animals at bothtime points (Figure 5C and 5D). Morphometric analysis ofserial sections showed a decrease of �58% in neointimalthickness in the G-CSF group at 2 (0.0148�0.0025, n�6) and4 (0.0171�0.0039, n�6) weeks after injury relative to salinecontrols (2 weeks, 0.0356�0.0034, n�6; 4 weeks,0.0412�0.0057, n�5, P�0.05; Figure 5E), Neointimalcross-sectional area decreased by 160% in the G-CSF groupat 2 (0.0182�0.0032, n�4) and 4 (0.0128�0.0037, n�5)weeks compared with the control animals (2 weeks,0.0475�0.0102, n�4; 4 weeks, 0.0341�0.005, n�5,P�0.05). Comparable changes were seen in neointima-mediaratios in the G-CSF–treated group (2 weeks, 0.195�0.035; 4weeks, 0.311�0.84) compared with the control group (2weeks, 0.506�0.069; 4 weeks, 0.656�0.074, P�0.05; Figure5F). Concomitant with the changes in neointimal thickness,there was a 9% to 10% increase in luminal diameter in theG-CSF–treated animals at both time points (2 weeks,0.765�0.023; 4 weeks, 0.749�0.021) compared with thesaline group (2 weeks, 0.691�0.011; 4 weeks,0.684�0.0049, P�0.05; Figure 5G). Luminal area was cor-respondingly increased in the G-CSF group by 23% at 2(0.461�0.028, n�4) and by 21% at 4 (0.334�0.0185, n�5)weeks relative to the saline controls (2 weeks, 0.376�0.012;4 weeks, 0.274�0.004, n�4, P�0.05), suggesting eccentricremodeling in the G-CSF–treated group. Medial thickness didnot differ between the G-CSF–treated and control animals ateither time point (Figure 5H).

DiscussionDisruption of the endothelium triggers a number of signalingcascades that converge on medial smooth muscle cells to

stimulate their proliferation and migration, leading to patho-logical repair and the development of neointimal hyperpla-sia.1,3,4 Although the injury itself may stimulate reendotheli-alization of the denuded vessel,2,4 the time required for thisnative repair process to restore endothelial function appearsbe too long to prevent the early critical events leading toactivation of vascular smooth muscle cells and neointimalhyperplasia. Accordingly, we postulated that a therapeuticapproach that could promote early reendothelialization of theinjured vessels would potentiate the endogenous repair pro-cess. In the present study, we used G-CSF pretreatment as astrategy to stimulate the mobilization of circulating putativeEPCs to enhance rapid reendothelization of balloon-injuredvessels. Our results show that stimulation with G-CSF in-creases the abundance of circulating mononuclear cells ex-pressing the endothelial lineage phenotype. More signifi-cantly, we demonstrate that G-CSF pretreatment acceleratesthe rate of reendothelialization and inhibits neointimal thick-ening in balloon-injured carotid arteries.

We and several other groups have recently reported thatautologous CEPCs can be harvested from peripheral bloodand transplanted into denuded vessels.6–9,17 However, celltransplantation protocols for vessel repair are time consumingand technically challenging. Moreover, the ability to obtain asufficient number of cells may be limited by the onset of cellsenescence.18,19 Here, we show that a noninvasive approachbased on a well-established protocol of cytokine-inducedmobilization of hematopoietic precursor cells, when admin-istered in advance of vascular injury, confers significantprotection against the subsequent development of neointimalhyperplasia. This appears to be, at least in part, due to theincreased availability of circulating progenitor cells capableof inducing rapid reendothelialization of the injured vessel

Figure 4. Reendothelialization of balloon-denuded carotid arteries 2 weeks afterinjury. A and B, Low-power-magnification photomicrograph of vesselprofiles stained with anti-CD31 andRECA-1 antibodies, respectively, in fro-zen cross sections from untreated ani-mals 14 days after injury. Arrows indicateminimal CD31-positive staining. C, Topo-graphic SEM view of untreated vessel.Arrows show incomplete and sparse en-dothelium. D and E, Low-power view ofCD31 and RECA-1 staining, respectively,of vessel profiles from G-CSF–treatedanimals. Arrows indicate continuousmonolayer staining positive for CD31 andRECA-1 lining lumen of vessel. F, SEMview of luminal surface in G-CSF–treatedvessel shows strings of densely packedendothelium-like cells (arrows) coveringluminal surface. F, Percentage of ECcoverage by planar morphometry.*P�0.05, saline vs G-CSF. Histologicalsections were viewed at �100. SEMsamples were viewed at �1000. Allabbreviations are as defined in text.

Kong et al Bone Marrow Mobilization and Neointima Formation 2043

by guest on November 13, 2013http://circ.ahajournals.org/Downloaded from

Figure 5. Neointimal hyperplasia in balloon-denuded carotid arteries. Van Gieson elastic staining of vessel profiles from untreated ani-mals at A, 2 weeks and B, 4 weeks after injury. Elastic staining of vessel profiles from G-CSF–treated animals at C, 2 weeks and D, 4weeks after injury. Sections were viewed at �100. Planar morphometric analysis of E, neointima thickness; F, neointima-media ratio; G,lumen diameter; and H, medial thickness in untreated and G-CSF–treated animals at 2 and 4 weeks after balloon injury. Significant dif-ferences were seen in neointimal thickness, neointima-media ratios, and lumen diameter between G-CSF–treated and control animalsat 2 and 4 weeks after injury. *P�0.05, saline vs G-CSF. All abbreviations are as defined in text.

2044 Circulation October 5, 2004

by guest on November 13, 2013http://circ.ahajournals.org/Downloaded from

and preventing the activation of medial smooth muscle. Thisis suggested by the accelerated rate of reendothelializationand the accompanying reduction in inflammation in theinjured vessels of the G-CSF–treated animals. We hypothe-size that the rapid reconstitution of the denuded vessel by themobilized CEPCs leads to timely restoration of endothelialfunction and vascular homeostasis, resulting in inhibition ofneointimal formation.

We note that several groups have reported mobilization ofEPCs by cytokine treatment.9–13 Kocher et al11 reported thatstimulation with C-CSF increases the number of CD34-positive cells expressing endothelial markers. Also, usingG-CSF treatment, Bhattacharya et al12 and Shi et al13 showedenhanced endothelialization of small-caliber prosthetic graftsin association with an elevation in CEPCs. Our presentfindings provide further evidence of the therapeutic potentialof cytokine-induced mobilization of progenitor cells in vesselrepair. Specifically, we demonstrate for the first time thetherapeutic benefit of G-CSF pretreatment as a strategy forprevention of restenosis after angioplasty-induced vascularinjury. Others have reported that statin therapy enhancesreendothelialization and reduces neointimal formation ininjured vessels,14 apparently due to sustained mobilization ofCEPCs from the bone marrow. However, the mechanism bywhich statins mobilize CEPCs may differ from that of G-CSFand other cytokines. The effect of G-CSF on CEPC mobili-zation could be seen in the short term, whereas the effect ofstatin therapy may require several weeks of treatment. Thisdifference in time course may have therapeutic implications,in that the timing of administration and response could differ,especially in association with angioplasty and revasculariza-tion procedures.

Our study did not specifically examine the sources and themechanisms governing the mobilization, homing, and differ-entiation of CEPCs and their role in endothelial repair at thesite of injury. The bone marrow has been reported to be theprimary site of origin of progenitor cells5,6,9,12; however,contributions from other hematopoietic and nonhematopoi-etic tissues to the circulating progenitor cell pool cannot becompletely excluded. In addition, resident EPCs and nativeECs may also contribute to vascular repair. In the presentstudy, we eliminated the spleen as a source of CEPCs20 bysplenectomizing the animals before cytokine administration.Regarding the potential mechanisms involved in mobilizationand homing of the CEPCs, several groups have reported thatthe cells are recruited predominantly to sites of injury,7,11,14

suggesting that signals emanating from the injury site mayplay a role in the mobilization, homing, and differentiationprocesses. In addition, G-CSF may stimulate the migrationand proliferation of resident EPCs and ECs in the vicinity ofthe injury site.16 From a therapeutic standpoint, our resultsmay have clinical implications for the treatment of vasculo-proliferative disease. Mobilization of progenitor cells inadvance of angioplasty may provide a strategy for inhibitionof restenosis in injured vessels. This may represent a novelantirestenosis paradigm that focuses on disease prevention byharnessing the therapeutic potential of the native repair andregeneration processes. The simplicity and cost-effectivenessof this approach would be major advantages compared with

the stent and drug-based therapies currently in use. However,our observations are based on a splenectomized animalmodel. Clearly, the clinical feasibility of this approach willneed to be evaluated in an intact animal model of vasculo-proliferative disease. Despite this limitation, our resultsclearly show a significant therapeutic effect of G-CSF treat-ment in reendothelialization of injured blood vessels. On theother hand, the potential stimulation of inflammatory cells byG-CSF needs to be carefully evaluated. Recent evidencesuggests that G-CSF exerts counterbalancing effects on in-flammation by promoting neutrophilia while inhibiting proin-flammatory cytokine production.21 In addition, there is thepossibility that mobilized bone marrow progenitors maydifferentiate into vascular smooth muscle cells,22 which couldpotentially aggravate the severity of restenosis of treatedvessels. Clearly, the long-term outcome of this strategy andits safety and feasibility for use in patients will have to beestablished before the complete therapeutic potential of thisapproach can be assessed.

In conclusion, the present study shows that bone marrowstimulation with G-CSF before balloon-induced vascularinjury significantly increases the availability of CEPCs andaccelerates the rate of reendothelialization of injured vessels,leading to marked inhibition of neointimal formation andfavorable vascular remodeling. These findings suggest thatpretreatment with G-CSF may be a feasible and efficienttherapeutic strategy for prevention of restenosis after revas-cularization procedures, such as such as percutaneous trans-luminal angioplasty, stenting, and atherectomy.

AcknowledgmentsThis work was supported by National Institutes of Health grantsHL35610, HL058516, HL072010, and HL073219 to Dr Dzau and bygrants from the Canadian Institutes of Health Research and the Heartand Stroke Foundation of Saskatchewan to Dr Melo. Dr Melo is aNew Investigator of the Heart and Stroke Foundation of Canada andCanada Research Chair in Molecular Cardiology. Dr Gnecchi is arecipient of a scholarship from the Italian Society of Cardiology anda research award from the University of Pavia and IRCCS PoliclinicoSan Matteo, Pavia, Italy.

References1. Pauleto P, Sartore S, Pessina AC. Smooth muscle proliferation and

differentiation in neointima formation and vascular restenosis. Clin Sci.1994;87:467–479.

2. Schwartz RS. Pathophysiology of restenosis: interaction of thrombosis,hyperplasia and/or remodeling. Am J Cardiol. 1998;81:14E–17E.

3. Cines DB, Pollak ES, Buck CA, et al. Endothelial cells in physiology andin the pathophysiology of vascular disorders. Blood. 1998;91:3527–3561.

4. Behrendt D, Ganz P. Endothelial function: from vascular biology toclinical applications. Am J Cardiol. 2002;90 (suppl):40L–48L.

5. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitorendothelial cells for angiogenesis. Science. 1997;275:964–967.

6. Asahara T, Masuda H, Takahashi T, et al. Bone marrow origin ofendothelial progenitor cells responsible for postnatal vasculogenesis inphysiological and pathological neovascularization. Circ Res. 1999;85:221–228.

7. Kawamoto A, Gwon HC, Iwaguro H, et al. Therapeutic potential of exvivo expanded endothelial progenitor cells for myocardial ischemia. Cir-culation. 2001;103:634–637.

8. Griese DP, Ehsan A, Melo LG, et al. Isolation and transplantation ofautologous circulating endothelial cells into denuded vessels and pros-thetic grafts: implications for cell-based vascular therapy. Circulation.2003;108:2710–2715.

Kong et al Bone Marrow Mobilization and Neointima Formation 2045

by guest on November 13, 2013http://circ.ahajournals.org/Downloaded from

9. Kaushal S, Amiel GE, Gulserian KJ, et al. Functional small diameterneovessels created using endothelial progenitor cells expanded ex vivo.Nat Med. 2001;7:1035–1040.

10. Kalka C, Masuda H, Takahashi T, et al. Vascular endothelial growthfactor165 gene transfer augments circulating endothelial progenitor cells inhuman subjects. Circ Res. 2000;86:1198–1202.

11. Kocher AA, Schuster MD, Szabolcs MJ, et al. Neovascularization ofischemic myocardium by human bone-marrow-derived angioblastsprevents cardiomyocyte apoptosis, reduces remodeling and improvescardiac function. Nat Med. 2001;7:430–436.

12. Bhattacharya V, Shi Q, Ishida A, et al. Administration of granulocytecolony-stimulating factor enhances endothelialization and microvesselformation in small caliber synthetic vascular grafts. J Vasc Surg. 2000;32:116–123.

13. Shi Q, Bhattacharya V, Hong-D, et al. Utilizing granulocyte colony-stimulating factor to enhance vascular graft endothelialization from cir-culating blood cells. Ann Vasc Surg. 2002;16:314–320.

14. Walter DH, Rittig K, Bahlmann FH, et al. Statin therapy acceleratesreendothelialization: a novel effect involving mobilization and incorpo-ration of bone marrow-derived endothelial progenitor cells. Circulation.2002;105:3017–3024.

15. Lapidot T, Petit I. Current understanding of stem cell mobilization: theroles of chemokines, proteolytic enzymes, adhesion molecules, cytokines,and stromal cells. Exp Hematol. 2002;30:973–981.

16. Bussolino F, Wang JM, Defilippi P, et al. Granulocyte and granulocyte-macrophage-colony stimulating factors induce human endothelial cells tomigrate and proliferate. Nature. 1989;337:471–473.

17. Iwaguro H, Yamaguchi J, Kalka C, et al. Endothelial progenitor cellvascular endothelial growth factor gene transfer for vascular regeneration.Circulation. 2002;105:732–738.

18. Murasawa S, Llevadot J, Silver M, et al. Constitutive human telomerasereverse transcriptase expression enhances regenerative properties of en-dothelial progenitor cells. Circulation. 2002;106:1133–1139.

19. Tisdale JF, Hanazono Y, Sellers SE, et al. Ex vivo expansion of genet-ically marked rhesus peripheral blood progenitor cells results indiminished long-term repopulating ability. Blood. 1998;92:1131–1141.

20. Werner N, Junk S, Laufs U, et al. Intravenous transfusion of endothelialprogenitor cells reduces neointima formation after vascular injury. CircRes. 2003;93:e17–e24.

21. Hartung T. Anti-inflammatory effects of granulocyte colony-stimulatingfactor. Curr Opin Hematol. 1998;5:221–225.

22. Simper D, Stalboerger PG, Panetta CJ, et al. Smooth muscle progenitorcells in human blood. Circulation. 2002;106:1199 –1204.

2046 Circulation October 5, 2004

by guest on November 13, 2013http://circ.ahajournals.org/Downloaded from


Recommended